1
|
Zhang K, Zhang H, Wang B, Gao S, Sun C, Jia C, Cui J. NR2F1 overexpression alleviates trophoblast cell dysfunction by inhibiting GDF15/MAPK axis in preeclampsia. Hum Cell 2024; 37:1405-1420. [PMID: 39007956 DOI: 10.1007/s13577-024-01095-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 06/15/2024] [Indexed: 07/16/2024]
Abstract
Abnormal functions of trophoblast cells are associated with the pathogenesis of preeclampsia (PE). Nuclear receptor subfamily 2 group F member 1 (NR2F1) acts as a transcriptionally regulator in many diseases, but its role in PE remains unknown. Hypoxia/reoxygenation (H/R)-stimulated HTR-8/SVneo cells were used to mimic PE injury in vitro. NR2F1 overexpression alleviated trophoblast apoptosis, as evidenced by the decreased number of TUNEL-positive cells and the downregulation of caspase 3 and caspase 9 expression in cells. NR2F1 overexpression increased the invasion and migration ability of HTR-8/SVneo cells, accompanied by increased protein levels of matrix metalloproteinase (MMP)-2 and MMP-9. mRNA-seq was applied to explore the underlying mechanism of NR2F1, identifying growth differentiation factor 15 (GDF15) as the possible downstream effector. Dual-luciferase reporter, ChIP-qPCR, and DNA pull-down assays confirmed that NR2F1 bound to the promoter of GDF15 and transcriptionally inhibited its expression. GDF15 overexpression increased apoptosis and decreased the ability of invasion and migration in HTR-8/SVneo cells expressing NR2F1. MAPK pathway was involved in the regulation of PE. Administration of p38 inhibitor, ERK inhibitor, and JNK inhibitor reversed the effect of simultaneous overexpression NR2F1 and GDF15 on trophoblast apoptosis, invasion, and migration. Our findings demonstrated that NR2F1 overexpression inhibited trophoblast apoptosis and promoted trophoblast invasion and migration. NR2F1 might negatively regulate GDF15 expression by binding to its promoter region, which further inhibited MAPK signaling pathway in PE. Our study highlights that NR2F1 might sever as a potential target in PE.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Hailing Zhang
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Bing Wang
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Shanshan Gao
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Caiping Sun
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Cong Jia
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Jinquan Cui
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhengzhou University, No. 2, Jingba Road, Zhengzhou, Henan, People's Republic of China.
| |
Collapse
|
2
|
Gan X, Liu H, Chen D, Liu Z, Lu Q, Lai X, Hou H, Zhang M, Zhang JY, Duan Y, Lu S, Chen M, Lash GE, Ning F. Interleukin-1 beta signals through the ERK signalling pathway to modulate human placental trophoblast migration and invasion in the first trimester of pregnancy. Placenta 2024; 151:67-78. [PMID: 38723477 DOI: 10.1016/j.placenta.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/31/2024] [Accepted: 04/24/2024] [Indexed: 06/03/2024]
Abstract
INTRODUCTION Interleukin-1 beta (IL-1β) can promote cell migration, invasion and metastasis in various cancer cells. The mechanism of its role in human trophoblast has not been fully investigated. Therefore, we aimed to investigate the expression level of IL-1β in first trimester decidua and placenta and its potential role in regulation of extravillous trophoblast cell (EVT) invasion and migration. METHODS First trimester placenta and decidua were collected to study the expression levels of IL-1β and its receptors by immunohistochemical staining. Primary isolates of first trimester EVT or the HTR-8/SVneo trophoblast like cell line were used to assess migration and invasion. Matrix metalloproteinase levels were assessed by gelatin zymography and ELISA. The phosphorylation profile of signaling pathway proteins was detected with the Proteome Profiler Human Phospho-Kinase Array Kit. Differentially expressed proteins in cells was detected and verified by Western Blot. RESULTS IL-1β, its receptors and antagonist are expressed in first trimester placenta and decidua, exogenous IL-1β stimulates trophoblast cell outgrowth, migration and invasion through the ERK signaling pathway. IL-1β was significantly increased in the placenta at 6-7 weeks gestation compared with 8-9 weeks gestation (P < 0.0001). Transwell and RTCA assays indicated that IL-1β stimulates the invasion and migration of EVT. In addition, IL-1β promoted the phosphorylation of ERK 1/2. It also promoted the expression of MMP2 and MMP9 in EVT as demonstrated by gelatin zymography assay and enzyme linked immunosorbent assay. DISCUSSION This study demonstrated IL-1β expression in placenta and decidua, and that it regulates EVT invasion and migration.
Collapse
Affiliation(s)
- Xiaowen Gan
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, PR China
| | - Hanbo Liu
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, PR China
| | - Danyang Chen
- Cancer Research Institute, Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510623, PR China
| | - Zongcai Liu
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, PR China
| | - Qinsheng Lu
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, PR China
| | - Xingqiang Lai
- Organ Transplantation Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510623, PR China
| | - Huomei Hou
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, PR China
| | - Min Zhang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, PR China
| | - Joy Yue Zhang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, PR China
| | - Yaoyun Duan
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, PR China
| | - Shenjiao Lu
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, PR China
| | - Miaojuan Chen
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, PR China.
| | - Gendie E Lash
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, PR China.
| | - Fen Ning
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, PR China.
| |
Collapse
|
3
|
Liu W, Wang R, Liu S, Yin X, Huo Y, Zhang R, Li J. YKL-40 promotes proliferation and invasion of HTR-8/SVneo cells by activating akt/MMP9 signalling in placenta accreta spectrum disorders. J OBSTET GYNAECOL 2023; 43:2211681. [PMID: 37192383 DOI: 10.1080/01443615.2023.2211681] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/03/2023] [Indexed: 05/18/2023]
Abstract
YKL-40 is a secreted glycoprotein that can promote invasion, angiogenesis and inhibit apoptosis, and was highly expressed in a variety of tumours. In this paper, we investigated the impacts of YKL-40 on proliferation and invasion in HTR-8/SVneo cells during placenta accreta spectrum disorders (PAS) development. The levels of YKL-40 protein in late-pregnant placental tissue were detected using immunohistochemistry and Western blotting, and gene expression using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The proliferation, migration, invasion and apoptosis abilities of HTR-8/SVneo cells were detected by cell counting kit-8 (CCK-8), Transwell, scratch assay, and flow cytometry, respectively. Our current results showed that YKL-40 was significantly increased in the PAS group compared to the normal control group (P < 0.01). Biological function experiments showed that YKL-40 significantly promoted the proliferation, migration and invasion of HTR-8/SVneo cells, and inhibited cell apoptosis. Knockdown of YKL-40 inhibited the activation of Akt/MMP9 signalling in trophoblast cells. These data suggested that YKL-40 might be involved in the progression of PAS, which may be attributed to the regulation of Akt/MMP9 signalling pathway.
Collapse
Affiliation(s)
- Weifang Liu
- North China University of Science and Technology, Tangshan, China
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, China
| | - Runfang Wang
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, China
| | - Suxin Liu
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, China
| | - Xiaoqian Yin
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, China
| | - Yan Huo
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, China
| | - Ruiling Zhang
- North China University of Science and Technology, Tangshan, China
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, China
| | - Jia Li
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, China
- College of Postgraduate, Hebei North University, Zhangjiakou, China
| |
Collapse
|
4
|
Nurzadeh M, Ghalandarpoor-Attar SM, Ghalandarpoor-Attar SN, Rabiei M. The Role of Interferon (IFN)-γ in Extravillous Trophoblast Cell (EVT) Invasion and Preeclampsia Progression. Reprod Sci 2022; 30:1462-1469. [PMID: 36289172 DOI: 10.1007/s43032-022-01110-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 10/13/2022] [Indexed: 11/29/2022]
Abstract
The involvement of the immune system in pregnancy is a controversial subject. The functions of T helper (Th) 1 and Th2 cells have been proposed, that Th1 cytokines promoting allograft rejection may impair pregnancy, whereas Th2-type cytokines suppressing Th1 responses improve allograft tolerance and hence embryonic survival. Maternal-fetal tolerance begins in the uterus; therefore, optimal adaptation to the fetus is the result of a complex interference. The invasion of extravillous trophoblast cells (EVTs) into the decidua and the inner third of the myometrium is essential for a healthy pregnancy. The mechanisms that influence trophoblast invasion are unknown; however, cytokines from uterine natural killer (uNK) cells, NKT cells, macrophages, and T cells appear to be involved. All these cells are major sources of interferon gamma (IFN-γ). Recent studies have shown that IFN-γ can inhibit EVT invasion via a mechanism dependent on an increase in EVT apoptosis and a decrease in matrix metalloproteinases (MMPs). Regarding controversies in this context, this study aimed to comprehensively review the role of IFN-γ and IFN-γ-producing cells in EVT invasion, successful pregnancy, and preeclampsia.
Collapse
Affiliation(s)
- Maryam Nurzadeh
- Fetomaternal Department, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Maryam Rabiei
- Obstetrics and Gynecology Department, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Park JY, Mani S, Clair G, Olson HM, Paurus VL, Ansong CK, Blundell C, Young R, Kanter J, Gordon S, Yi AY, Mainigi M, Huh DD. A microphysiological model of human trophoblast invasion during implantation. Nat Commun 2022; 13:1252. [PMID: 35292627 PMCID: PMC8924260 DOI: 10.1038/s41467-022-28663-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 02/01/2022] [Indexed: 12/12/2022] Open
Abstract
Successful establishment of pregnancy requires adhesion of an embryo to the endometrium and subsequent invasion into the maternal tissue. Abnormalities in this critical process of implantation and placentation lead to many pregnancy complications. Here we present a microenigneered system to model a complex sequence of orchestrated multicellular events that plays an essential role in early pregnancy. Our implantation-on-a-chip is capable of reconstructing the three-dimensional structural organization of the maternal-fetal interface to model the invasion of specialized fetal extravillous trophoblasts into the maternal uterus. Using primary human cells isolated from clinical specimens, we demonstrate in vivo-like directional migration of extravillous trophoblasts towards a microengineered maternal vessel and their interactions with the endothelium necessary for vascular remodeling. Through parametric variation of the cellular microenvironment and proteomic analysis of microengineered tissues, we show the important role of decidualized stromal cells as a regulator of extravillous trophoblast migration. Furthermore, our study reveals previously unknown effects of pre-implantation maternal immune cells on extravillous trophoblast invasion. This work represents a significant advance in our ability to model early human pregnancy, and may enable the development of advanced in vitro platforms for basic and clinical research of human reproduction.
Collapse
Affiliation(s)
- Ju Young Park
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Sneha Mani
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Geremy Clair
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Heather M Olson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Vanessa L Paurus
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Charles K Ansong
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Cassidy Blundell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel Young
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica Kanter
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Scott Gordon
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alex Y Yi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Monica Mainigi
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Dan Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Rawlings TM, Makwana K, Tryfonos M, Lucas ES. Organoids to model the endometrium: implantation and beyond. REPRODUCTION AND FERTILITY 2022; 2:R85-R101. [PMID: 35118399 PMCID: PMC8801025 DOI: 10.1530/raf-21-0023] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/05/2021] [Indexed: 12/18/2022] Open
Abstract
Despite advances in assisted reproductive techniques in the 4 decades since the first human birth after in vitro fertilisation, 1–2% of couples experience recurrent implantation failure, and some will never achieve a successful pregnancy even in the absence of a confirmed dysfunction. Furthermore, 1–2% of couples who do conceive, either naturally or with assistance, will experience recurrent early loss of karyotypically normal pregnancies. In both cases, embryo-endometrial interaction is a clear candidate for exploration. The impossibility of studying implantation processes within the human body has necessitated the use of animal models and cell culture approaches. Recent advances in 3-dimensional modelling techniques, namely the advent of organoids, present an exciting opportunity to elucidate the unanswerable within human reproduction. In this review, we will explore the ontogeny of implantation modelling and propose a roadmap to application and discovery.
Collapse
Affiliation(s)
- Thomas M Rawlings
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Komal Makwana
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Maria Tryfonos
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Emma S Lucas
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK.,Centre for Early Life, Warwick Medical School, University of Warwick, Coventry, UK
| |
Collapse
|
7
|
Pu Y, Gingrich J, Veiga-Lopez A. A 3-dimensional microfluidic platform for modeling human extravillous trophoblast invasion and toxicological screening. LAB ON A CHIP 2021; 21:546-557. [PMID: 33166377 PMCID: PMC8212566 DOI: 10.1039/d0lc01013h] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Placental trophoblast cells invasion into the maternal uterus is an essential and complex event in the formation of the maternal-fetal interface. Commonly used two-dimensional (2D) cell invasion tools do not accurately represent the in vivo cell invasion microenvironment. Three-dimensional (3D) silicone polymer polydimethylsiloxane (PDMS) microfluidic platforms are an emerging technology in developing organ-on-a-chip models. Here, we present a placenta-on-a-chip platform that enables the evaluation of trophoblast invasion with intraluminal flow within an engineered PDMS 3D microfluidic chip. This platform reproduces key elements of the placental microenvironment, including endothelial and trophoblast cells, layered with an extracellular matrix, and incorporates dynamic medium flow while allowing for real-time monitoring, imaging, evaluation of trophoblast cell invasion, and heterocellular cell-to-cell interactions. Coupled with fluorescent cell tagging and flow cytometry, this platform also allows collection of the invasive cells. This will help our understanding of pathways that regulate trophoblast cell invasion and may prove important for toxicological screening of exposures that interfere with invasiveness in a complex organ such as the placenta.
Collapse
Affiliation(s)
- Yong Pu
- Department of Pathology, University of Illinois at Chicago, 909 S. Wolcott Ave, Rm 6093, Chicago, IL 60612, USA.
| | - Jeremy Gingrich
- Department of Pharmacology and Toxicology, Michigan State University, USA
| | - Almudena Veiga-Lopez
- Department of Pathology, University of Illinois at Chicago, 909 S. Wolcott Ave, Rm 6093, Chicago, IL 60612, USA.
| |
Collapse
|
8
|
Perfluoroctanoic acid (PFOA) enhances NOTCH-signaling in an angiogenesis model of placental trophoblast cells. Int J Hyg Environ Health 2020; 229:113566. [PMID: 32485599 DOI: 10.1016/j.ijheh.2020.113566] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/19/2020] [Accepted: 05/19/2020] [Indexed: 01/09/2023]
Abstract
Exposure to perfluoroalkyl substances (PFAS) was found to be associated with several pathological endpoints, including high cholesterol levels, specific defective functions of the immune system and reduced birth weight. While environmental PFAS have been recognized as threats for public health, surprisingly little is known about the underlying mechanisms of toxicity. We hypothesized that some of the observed vascular and developmental effects of environmental PFAS may share a common molecular pathway. At elevated levels of exposure to PFAS, a reduction in mean birth weight of newborns has been observed in combination with a high incidence rate of preeclampsia. As both, preeclampsia and reduced birth weight are consequences of an inadequate placental vascularization, we hypothesized that the adaptation of placental vasculature may get compromised by PFAS. We analyzed pseudo-vascular network formation and protein expression in the HTR8/SVneo cell line, an embryonic trophoblast cell type that is able to form vessel-like vascular networks in 3D-matrices, similar to endothelial cells. PFOA (perfluoroctanoic acid), but not PFOS (perfuoroctanesulfonic acid), induced morphological changes in the vascular 3D-network structure, without indications of compromised cellular viability. Incubation with PFOA reduced cellular sprouting and elongated isolated stalks in pseudo-vascular networks, while a γ-secretase inhibitor BMS-906024 induced directional opposite effects. We found a PFOA-induced increase in NOTCH intracellular domain (NICD) abundance in HTR8/SVneo, indicating that PFOA enhances NOTCH-signaling in this cell type. Enhancement of NOTCH-pathway by PFOA may be a key to understand the mode of action of PFAS, as this pathway is critically involved in many confirmed physiological/toxicological symptoms associated with PFAS exposure.
Collapse
|
9
|
Dobaño C, Bardají A, Kochar S, Kochar SK, Padilla N, López M, Unger HW, Ome-Kaius M, Castellanos ME, Arévalo-Herrera M, Hans D, Martínez-Espinosa FE, Bôtto-Menezes C, Malheiros A, Desai M, Casellas A, Chitnis CE, Rogerson S, Mueller I, Menéndez C, Requena P. Blood cytokine, chemokine and growth factor profiling in a cohort of pregnant women from tropical countries. Cytokine 2019; 125:154818. [PMID: 31514106 DOI: 10.1016/j.cyto.2019.154818] [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: 05/13/2019] [Revised: 08/20/2019] [Accepted: 08/20/2019] [Indexed: 12/28/2022]
Abstract
The immune status of women changes during and after pregnancy, differs between blood compartments at delivery and is affected by environmental factors particularly in tropical areas endemic for multiple infections. We quantified the plasma concentration of a set of thirty-one TH1, TH2, TH17 and regulatory cytokines, pro-inflammatory and anti-inflammatory cytokines and chemokines, and growth factors (altogether biomarkers), in a cohort of 540 pregnant women from five malaria-endemic tropical countries. Samples were collected at recruitment (first antenatal visit), delivery (periphery, cord and placenta) and postpartum, allowing a longitudinal analysis. We found the lowest concentration of biomarkers at recruitment and the highest at postpartum, with few exceptions. Among them, IL-6, HGF and TGF-β had the highest levels at delivery, and even higher concentrations in the placenta compared to peripheral blood. Placental concentrations were generally higher than peripheral, except for eotaxin that was lower. We also compared plasma biomarker concentrations between the tropical cohort and a control group from Spain at delivery, presenting overall higher biomarker levels the tropical cohort, particularly pro-inflammatory cytokines and growth factors. Only IL-6 presented lower levels in the tropical group. Moreover, a principal component analysis of biomarker concentrations at delivery showed that women from Spain grouped more homogenously, and that IL-6 and IL-8 clustered together in the tropical cohort but not in the Spanish one. Plasma cytokine concentrations correlated with Plasmodium antibody levels at postpartum but not during pregnancy. This basal profiling of immune mediators over gestation and in different compartments at delivery is important to subsequently understand response to infections and clinical outcomes in mothers and infants in tropical areas.
Collapse
Affiliation(s)
- Carlota Dobaño
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer del Rosselló, 132, 08036 Barcelona, Spain.
| | - Azucena Bardají
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer del Rosselló, 132, 08036 Barcelona, Spain
| | - Swati Kochar
- Medical College, PBM Hospital, Bikaner, Rajasthan 334001, India
| | - Sanjay K Kochar
- Medical College, PBM Hospital, Bikaner, Rajasthan 334001, India
| | - Norma Padilla
- Centro de Estudios en Salud, Universidad del Valle de Guatemala, 18 Avenida 11-95, Guatemala 01015, Guatemala
| | - Marta López
- Department of Maternal-Fetal Medicine, Hospital Clínic-IDIBAPS, CIBER-ER, Carrer del Rosselló, 149, 08036 Barcelona, Spain
| | - Holger W Unger
- Papua New Guinea Institute of Medical Research, P.O. Box 378, Madang 511, Papua New Guinea
| | - Maria Ome-Kaius
- Papua New Guinea Institute of Medical Research, P.O. Box 378, Madang 511, Papua New Guinea
| | - Maria Eugenia Castellanos
- Centro de Estudios en Salud, Universidad del Valle de Guatemala, 18 Avenida 11-95, Guatemala 01015, Guatemala
| | | | - Dhiraj Hans
- International Center for Genetic Engineering and Biotechnology, Jawaharlal Nehru University, Aruna Asaf Ali Marg, New Delhi, Delhi 110067, India
| | - Flor E Martínez-Espinosa
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Av. Pedro Teixeira, s/n - Dom Pedro, Manaus, AM 69040-000, Brazil; Instituto Leônidas e Maria Deane, Rua Teresina, 476 - Adrianópolis, Manaus 69.057-070, Brazil
| | - Camila Bôtto-Menezes
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Av. Pedro Teixeira, s/n - Dom Pedro, Manaus, AM 69040-000, Brazil; Universidade do Estado do Amazonas, 69850-000, R. Bloco Um e Três, 4-40 - Platô do Piquiá, Boca do Acre, AM 69850-000, Brazil
| | - Adriana Malheiros
- Instituto de Ciências Biológicas, Universidade Federal do Amazonas, Av. Jauary Marinho - Setor Sul - Coroado, Manaus, AM, Brazil
| | - Meghna Desai
- Centers for Disease Control and Prevention, Division of Parasitic Diseases and Malaria, Malaria Branch, 1600 Clifton Rd, Atlanta, GA 30333, USA
| | - Aina Casellas
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer del Rosselló, 132, 08036 Barcelona, Spain
| | - Chetan E Chitnis
- International Center for Genetic Engineering and Biotechnology, Jawaharlal Nehru University, Aruna Asaf Ali Marg, New Delhi, Delhi 110067, India; Malaria Parasite Biology and Vaccines Unit, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | | | - Ivo Mueller
- Walter and Eliza Hall Institute, 1G, Royal Parade, Parkville, VIC 3052, Australia
| | - Clara Menéndez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer del Rosselló, 132, 08036 Barcelona, Spain
| | - Pilar Requena
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer del Rosselló, 132, 08036 Barcelona, Spain; Departmento de Medicina Preventiva y Salud Pública, Universidad de Granada, Facultad de Farmacia, Campus de Cartuja, 18071 Granada, Spain.
| |
Collapse
|
10
|
Beltrame JS, Scotti L, Sordelli MS, Cañumil VA, Franchi AM, Parborell F, Ribeiro ML. Lysophosphatidic acid induces the crosstalk between the endovascular human trophoblast and endothelial cells in vitro. J Cell Physiol 2019; 234:6274-6285. [PMID: 30362520 DOI: 10.1002/jcp.27358] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 08/17/2018] [Indexed: 12/29/2022]
Abstract
Spiral artery remodeling at the maternal-fetal interface is crucial for successful pregnancy and requires the interaction between the first trimester trophoblast and the endothelial cells of the maternal vessels. However, the precise mechanism of this dialog has yet to be determined. The current study investigated whether lysophosphatidic acid (LPA) modulates trophoblast-endothelial crosstalk in vitro. HTR-8/SVneo trophoblast cell line (H8) was seeded on top of Geltrex, incubated with LPA or LPA + NS-398 (selective cyclooxygenase-2 inhibitor), LPA + 1400W (selective inducible nitric oxide synthase inhibitor) or LPA + IL-6 neutralizing antibody and assayed for tube formation to model the acquisition of trophoblast endovascular phenotype. The supernatants were collected and used as conditioned media (CM). To test trophoblast-endothelial crosstalk, the endothelial cell line EA.hy926 was incubated with trophoblast CM. The CM from LPA-induced tubulogenesis stimulated endothelial cells migration and did not modify the apoptosis. Soluble factors derived from cyclooxygenase-2 and IL-6 pathways were involved in H8-EA.hy926 interaction under the LPA effect. Moreover, LPA increased the levels of IL-6 mRNA by cyclooxygenase-2 pathway in H8 cells. Collectively, LPA promotes trophoblast-endothelial crosstalk in vitro and induces the release of trophoblast soluble factors that stimulate endothelial cells migration without changes in apoptosis. The evidence presented here provides new insights about an active role of LPA as a lipid mediator regulating vascular remodeling at the maternal-fetal interface.
Collapse
Affiliation(s)
- Jimena S Beltrame
- Laboratorio de Fisiología y Farmacología de la Reproducción, Centro de Estudios Farmacológicos y Botánicos (CEFyBO) (CONICET - Facultad de Medicina, Universidad de Buenos Aires), Paraguay 2155, 16th floor, Buenos Aires, Argentina
| | - Leopoldina Scotti
- Laboratorio de Estudios de la Fisiopatología del Ovario, Instituto de Biología y Medicina Experimental (IByME) - (CONICET), Vuelta de Obligado 2490, Buenos Aires, Argentina
| | - Micaela S Sordelli
- Laboratorio de Fisiología y Farmacología de la Reproducción, Centro de Estudios Farmacológicos y Botánicos (CEFyBO) (CONICET - Facultad de Medicina, Universidad de Buenos Aires), Paraguay 2155, 16th floor, Buenos Aires, Argentina
| | - Vanesa A Cañumil
- Laboratorio de Fisiología y Farmacología de la Reproducción, Centro de Estudios Farmacológicos y Botánicos (CEFyBO) (CONICET - Facultad de Medicina, Universidad de Buenos Aires), Paraguay 2155, 16th floor, Buenos Aires, Argentina
| | - Ana M Franchi
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos (CEFyBO) (CONICET - Facultad de Medicina, Universidad de Buenos Aires), Paraguay 2155, 16th floor, Buenos Aires, Argentina
| | - Fernanda Parborell
- Laboratorio de Estudios de la Fisiopatología del Ovario, Instituto de Biología y Medicina Experimental (IByME) - (CONICET), Vuelta de Obligado 2490, Buenos Aires, Argentina
| | - María L Ribeiro
- Laboratorio de Fisiología y Farmacología de la Reproducción, Centro de Estudios Farmacológicos y Botánicos (CEFyBO) (CONICET - Facultad de Medicina, Universidad de Buenos Aires), Paraguay 2155, 16th floor, Buenos Aires, Argentina
| |
Collapse
|
11
|
Xu Y, Sui L, Qiu B, Yin X, Liu J, Zhang X. ANXA4 promotes trophoblast invasion via the PI3K/Akt/eNOS pathway in preeclampsia. Am J Physiol Cell Physiol 2019; 316:C481-C491. [PMID: 30673304 DOI: 10.1152/ajpcell.00404.2018] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The inadequate trophoblast invasion is associated with the development of preeclampsia (PE). Considering that annexin A4 (ANXA4) enhances tumor invasion, we aimed to explore the functional role of ANXA4 in trophoblast cells and to examine the underlying mechanism. ANXA4 expression in PE placentas was analyzed using immunohistochemistry and Western blotting. Cell proliferation, invasion, and apoptosis were determined using a MTT assay, Transwell assay, and flow cytometry, respectively. The expression levels of matrix metalloproteinase (MMP)-2, MMP-9, phosphoinositide 3-kinase (PI3K), Akt, phosphorylated (p)-Akt, and phosphorylated endothelial nitric oxide synthase (p-eNOS) were detected by Western blotting. Placentas were prepared for pathological examination using hematoxylin and eosin staining and apoptosis determination using the TUNEL method. Expression of ANXA4, PI3K, p-Akt and p-eNOS was downregulated in human PE placentas and PE placenta-derived extravillous cytotrophoblasts (EVCTs). Furthermore, ANXA4 overexpression promoted cell proliferation and invasion, inhibited cell apoptosis, and upregulated protein expression of PI3K, p-Akt, and p-eNOS in human trophoblast cells HTR-8/SVneo and JEG-3. By contrast, ANXA4 knockdown exerted the opposite effects. Furthermore, inhibition of the PI3K/Akt pathway by LY294002 abrogated the ANXA4 overexpression-mediated effects on trophoblast behavior. Furthermore, eNOS knockdown abrogated the ANXA4 overexpression-induced promotion of cell invasion and MMP2/9 expression. Additionally, in N-nitro-l-arginine methyl ester (l-NAME)-induced PE rats, ANXA4 overexpression alleviated PE progression, accompanied by an increase in expression of PI3K, p-Akt, and p-eNOS in rat placentas. Our findings demonstrate that ANXA4 expression is downregulated in PE. ANXA4 may promote trophoblast invasion via the PI3K/Akt/eNOS pathway.
Collapse
Affiliation(s)
- Yalan Xu
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| | - Lili Sui
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, China
| | - Bintao Qiu
- Central Laboratory, Peking Union Medical College Hospital, Beijing, China
| | - Xiuju Yin
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| | - Juntao Liu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, China
| | - Xiaohong Zhang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
12
|
Weiss G, Sundl M, Glasner A, Huppertz B, Moser G. The trophoblast plug during early pregnancy: a deeper insight. Histochem Cell Biol 2016; 146:749-756. [PMID: 27510415 PMCID: PMC5101277 DOI: 10.1007/s00418-016-1474-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2016] [Indexed: 11/24/2022]
Abstract
During the first trimester of pregnancy, foetal endovascular trophoblasts invade into maternal spiral arteries, accumulate and form plugs in the lumen of the vessels. These plugs only allow blood plasma to seep through. Hence, during the first trimester of pregnancy, a first flow of fluids through the placental intervillous space is established, resulting in a physiological oxygen gradient between mother and foetus. The trophoblast plugs block spiral arteries until the beginning of the second trimester (11-14 weeks). In parallel, uterine glands are invaded and opened by endoglandular trophoblasts towards the intervillous space of the placenta, without showing the formation of plugs (Moser et al. in Hum Reprod 25:1127-1136, 2010, Hum Reprod Oxf Engl 30:2747-2757, 2015). This enables histiotrophic nutrition of the embryo prior to onset of maternal blood flow into the placenta. Failure of these endovascular and endoglandular invasion processes may lead to miscarriage or pregnancy disorders such as intrauterine growth restriction (IUGR). After dissolution of the plugs, the onset of maternal blood flow allows maternal blood cells to enter the intervillous space and oxygen concentrations rise up. In this study, we demonstrate for the first time serial cross sections through a trophoblast plug in a first trimester placental bed specimen. Invaded and plugged arteries as well as invaded uterine glands in week 11 of gestation are visualized with specific immunohistochemical double staining techniques. We show that spiral artery plugs appear throughout the placental invasion zone and illustrate erythrocytes stowed due to trophoblast plugs. In addition, we give evidence of the presence of MMP-1 in plugs of invaded spiral arteries. The results reveal a better understanding and a closer insight into the morphological appearance of trophoblast plugs and the consequences for placental and uterine blood flow.
Collapse
Affiliation(s)
- Gregor Weiss
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Harrachgasse 21/7, 8010, Graz, Austria
| | - Monika Sundl
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Harrachgasse 21/7, 8010, Graz, Austria
| | | | - Berthold Huppertz
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Harrachgasse 21/7, 8010, Graz, Austria
| | - Gerit Moser
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Harrachgasse 21/7, 8010, Graz, Austria.
| |
Collapse
|