1
|
Lapehn S, Nair S, Firsick EJ, MacDonald J, Thoreson C, Litch JA, Bush NR, Kadam L, Girard S, Myatt L, Prasad B, Sathyanarayana S, Paquette AG. A transcriptomic comparison of in vitro models of the human placenta. Placenta 2025; 159:52-61. [PMID: 39637677 DOI: 10.1016/j.placenta.2024.11.007] [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: 07/12/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Selecting an in vitro culture model of the human placenta is challenging due to representation of different trophoblast cell types with distinct biological roles and limited comparative studies that define key characteristics of these models. The aim of this research was to compare the transcriptomes of common in vitro models of the human placenta compared to bulk human placental tissue. METHODS We performed differential gene expression analysis on publicly available transcriptomic data from 7 in vitro models of the human placenta (HTR-8/SVneo, BeWo, JEG-3, JAR, Primary Trophoblasts, Villous Explants, and Trophoblast Stem Cells) and compared to bulk placental tissue from 2 cohort studies (CANDLE and GAPPS) or individual trophoblast cell types derived from bulk placental tissue. RESULTS All in vitro placental models had a substantial number of differentially expressed genes (DEGs, FDR<0.01) compared to the CANDLE and GAPPS placentas (Average DEGs = 10,624), and the individual trophoblast cell types (Average DEGs = 5413), indicating that there are vast differences in gene expression. Hierarchical clustering identified 54 gene clusters with distinct expression profiles across placental models, with 23 clusters enriched for specific KEGG pathways. Placental cell lines were classified by fetal sex based on expression of Y-chromosome genes that identified HTR-8/SVneo cells as female origin, while JEG-3, JAR, and BeWo cells are of male origin. DISCUSSION None of the models were a close approximation of the human bulk placental transcriptome, highlighting the challenges with model selection. To enable appropriate model selection, we adapted our data into a web application: "Comparative Transcriptomic Placental Model Atlas (CTPMA)".
Collapse
Affiliation(s)
- Samantha Lapehn
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA.
| | - Sidharth Nair
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Evan J Firsick
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA, 98195, USA
| | - Ciara Thoreson
- Global Alliance to Prevent Prematurity and Stillbirth, Lynwood, WA, 98036, USA
| | - James A Litch
- Global Alliance to Prevent Prematurity and Stillbirth, Lynwood, WA, 98036, USA
| | - Nicole R Bush
- Department of Psychiatry and Behavioral Sciences, Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Leena Kadam
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Sheela Sathyanarayana
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA; Center for Child Health, Behavior and Development, Seattle Children's Research Institute, Seattle, WA, 98101, USA; Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, 98101, USA
| | - Alison G Paquette
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA; Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA, 98195, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA
| |
Collapse
|
2
|
Dimova T, Alexandrova M, Vangelov I, You Y, Mor G. The modeling of human implantation and early placentation: achievements and perspectives. Hum Reprod Update 2024:dmae033. [PMID: 39673726 DOI: 10.1093/humupd/dmae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 10/29/2024] [Indexed: 12/16/2024] Open
Abstract
BACKGROUND Successful implantation is a critical step for embryo survival. The major losses in natural and assisted human reproduction appeared to occur during the peri-implantation period. Because of ethical constraints, the fascinating maternal-fetal crosstalk during human implantation is difficult to study and thus, the possibility for clinical intervention is still limited. OBJECTIVE AND RATIONALE This review highlights some features of human implantation as a unique, ineffective and difficult-to-model process and summarizes the pros and cons of the most used in vivo, ex vivo and in vitro models. We point out the variety of cell line-derived models and how these data are corroborated by well-defined primary cells of the same nature. Important aspects related to the handling, standardization, validation, and modus operandi of the advanced 3D in vitro models are widely discussed. Special attention is paid to blastocyst-like models recapitulating the hybrid phenotype and HLA profile of extravillous trophoblasts, which are a unique yet poorly understood population with a major role in the successful implantation and immune mother-embryo recognition. Despite raising new ethical dilemmas, extended embryo cultures and synthetic embryo models are also in the scope of our review. SEARCH METHODS We searched the electronic database PubMed from inception until March 2024 by using a multi-stage search strategy of MeSH terms and keywords. In addition, we conducted a forward and backward reference search of authors mentioned in selected articles. OUTCOMES Primates and rodents are valuable in vivo models for human implantation research. However, the deep interstitial, glandular, and endovascular invasion accompanied by a range of human-specific factors responsible for the survival of the fetus determines the uniqueness of the human implantation and limits the cross-species extrapolation of the data. The ex vivo models are short-term cultures, not relevant to the period of implantation, and difficult to standardize. Moreover, the access to tissues from elective terminations of pregnancy raises ethical and legal concerns. Easy-to-culture cancer cell lines have many limitations such as being prone to spontaneous transformation and lacking decent tissue characteristics. The replacement of the original human explants, primary cells or cancer cell lines with cultures of immortalized cell lines with preserved stem cell characteristics appears to be superior for in vitro modeling of human implantation and early placentation. Remarkable advances in our understanding of the peri-implantation stages have also been made by advanced three dimensional (3D) models i.e. spheroids, organoids, and assembloids, as placental and endometrial surrogates. Much work remains to be done for the optimization and standardization of these integrated and complex models. The inclusion of immune components in these models would be an asset to delineate mechanisms of immune tolerance. Stem cell-based embryo-like models and surplus IVF embryos for research bring intriguing possibilities and are thought to be the trend for the next decade for in vitro modeling of human implantation and early embryogenesis. Along with this research, new ethical dilemmas such as the moral status of the human embryo and the potential exploitation of women consenting to donate their spare embryos have emerged. The careful appraisal and development of national legal and ethical frameworks are crucial for better regulation of studies using human embryos and embryoids to reach the potential benefits for human reproduction. WIDER IMPLICATIONS We believe that our data provide a systematization of the available information on the modeling of human implantation and early placentation and will facilitate further research in this field. A strict classification of the advanced 3D models with their pros, cons, applicability, and availability would help improve the research quality to provide reliable outputs.
Collapse
Affiliation(s)
- Tanya Dimova
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Marina Alexandrova
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Ivaylo Vangelov
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Yuan You
- C.S. Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| |
Collapse
|
3
|
Su H, Li M, Li N, Zhang Y, He Y, Zhang Z, Zhang Y, Gao Q, Xu Z, Tang J. Endothelin-1 potentiated constriction in preeclampsia placental veins: Role of ETAR/ETBR/CaV1.2/CALD1. Placenta 2024; 158:165-174. [PMID: 39476475 DOI: 10.1016/j.placenta.2024.10.011] [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: 05/22/2024] [Revised: 10/11/2024] [Accepted: 10/20/2024] [Indexed: 12/11/2024]
Abstract
BACKGROUND Placenta plays a vital role in preeclampsia. The present study investigated the role of endothelin-1 (ET-1) and its receptors in preeclampsia placenta. METHOD Placenta samples were collected from normal and preeclampsia pregnancies, with one single fetus. Placental chorionic plate vessel tone was measured with DMT using vasoactive agents with or without antagonists. Role of L-type voltage-dependent calcium channels (CaV1.2) in single smooth muscle cell was detected using whole-cell patch clamp. PCR, Western blot, and ELISA was used to detect molecule expressions. Placental vessel explants and human umbilical vein smooth muscle cell (HUVSMC) were exposed to ET-1 treatment with or without antagonists. Human umbilical vein endothelial cell (HUVEC) and pregnant sheep was exposed to hypoxic condition, simulating preeclampsia. RESULTS ET-1 and IRL1620 mediated stronger contractions in preeclampsia placental veins, despite unchanged ETAR and decreased ETBR expression. Comparing with control, there was higher ET-1 in umbilical plasma, maternal plasma, and placental vessels from preeclampsia. In utero hypoxia increased plasma ET-1 in fetal lambs and ewes. Hypoxia promoted ET-1 production in HUVEC. Role and expression of CaV1.2 was decreased in preeclampsia placental vessels, while high-molecular-weight caldesmon (CALD1), the marker of contractile phenotype of smooth muscle cells, was significantly increased. ET-1 treatment increased CALD1 in placental explants and in HUVSMC via ETAR/ETBR. CONCLUSION The present study firstly demonstrated ET-1 induced greater contraction in preeclampsia placental chorionic plate veins via ETAR/ETBR, instead of via weaker CaV1.2. In utero hypoxia promoted plasma ET-1 in fetal lambs and ewe, similar to that in preeclampsia. ET-1, binding with ETAR/ETBR increased CALD1, which was associated with stronger contraction in preeclampsia. The data provided important information in preeclampsia onset.
Collapse
Affiliation(s)
- Hongyu Su
- Institute for Fetology, The First Affiliated Hospital of Soochow University, China
| | - Min Li
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Soochow University, China
| | - Na Li
- Perinatology Laboratory, Maternity and Child Health Care Hospital of Wuxi, China
| | - Yingying Zhang
- Perinatology Laboratory, Maternity and Child Health Care Hospital of Wuxi, China
| | - Yun He
- Department of Gynecology and Obstetrics, Taixing People's Hospital, China
| | - Ze Zhang
- Department of Gynecology and Obstetrics, Taixing People's Hospital, China
| | - Yumeng Zhang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, China
| | - Qinqin Gao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, China
| | - Zhice Xu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, China; Perinatology Laboratory, Maternity and Child Health Care Hospital of Wuxi, China
| | - Jiaqi Tang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, China.
| |
Collapse
|
4
|
Wang L, Zhang J, Sun A, Yin Y, Shen Y, Zhang D, Chen Q, Zhao M. Placental extracellular vesicles promoted cervical tumour tissue undergoing death. Placenta 2024; 158:223-230. [PMID: 39504594 DOI: 10.1016/j.placenta.2024.10.069] [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: 09/05/2024] [Revised: 10/15/2024] [Accepted: 10/30/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Cervical cancer is a leading cause of death in developing countries. Although the placenta is a tumor-like organ, the placental development, including invasive function, is well controlled. One mechanism is that extracellular vesicles (EVs) released from the placenta contribute to this regulation. Placental EVs carry functional proteins and regulatory RNAs. Our previous study reported that placental EVs inhibited ovarian cancer growth in vitro and in vivo. METHODS Whether the inhibitory effect induced by placental EVs also applies to cervical cancer, a non-endocrine-related cancer, in this study, we first co-cultured the cervical tumour tissues with placental explants. RESULTS Co-culturing cervical tumour tissues (n = 7) with placental explants showed necrotic signs and increased levels of senescence-associated proteins and death-associated miRNAs, including miRNA-143-3p, miRNA-519a-5p and miRNA-199a-3p in tumour tissues. Additionally, treatment of HeLa cells with placental EVs reduced the viability of HeLa cells and inhibited the ability of invasion and migration of HeLa cells. Increased levels of senescence-associated proteins and reduced levels of proliferative proteins may contribute to the inhibitory effects in HeLa cells. DISCUSSION placental EVs are involved in regulating placental development, and the delivery of cargo significantly impacts the functions of target cells. This study found that factors released from placental explants, likely placental EVs, had anti-tumour effects on the cervical tumour by inhibiting cervical cancer cell viability, invasion, and migration. Cargo in placental EVs, such as cellular death-associated miRNAs, may contribute to the inhibitory effects on cervical tumour.
Collapse
Affiliation(s)
- Lin Wang
- Department of Gynaecology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China; Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jinqiu Zhang
- Department of Pathology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Angang Sun
- Department of Pathology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yongxiang Yin
- Department of Pathology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Ye Shen
- Department of Family Planning, Wuxi Maternity and Child Care Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Dengxin Zhang
- Department of Anesthesia, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Qi Chen
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| | - Min Zhao
- Department of Gynaecology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China.
| |
Collapse
|
5
|
de Sousa AKA, Pires KSN, Cavalcante IH, Cavalcante ICL, Santos JD, Queiroz MIC, Leite ACR, Crispim AC, da Rocha Junior ER, Aquino TM, Weingrill RB, Urschitz J, Ospina-Prieto S, Borbely AU. Polystyrene microplastics exposition on human placental explants induces time-dependent cytotoxicity, oxidative stress and metabolic alterations. Front Endocrinol (Lausanne) 2024; 15:1481014. [PMID: 39634179 PMCID: PMC11614646 DOI: 10.3389/fendo.2024.1481014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Microplastics (MPs) are environmental pollutants that pose potential risks to living organisms. MPs have been shown to accumulate in human organs, including the placenta. In this study, we investigated the biochemical impact of 5 μm polystyrene microplastics (PS-MPs) on term placental chorionic villi explants, focusing on cytotoxicity, oxidative stress, metabolic changes, and the potential for MPs to cross the placental barrier. Methods Term placental chorionic explants were cultured for 24 hours with varying concentrations of PS-MPs, with MTT assays used to determine the appropriate concentration for further analysis. Cytotoxicity was assessed using the lactate dehydrogenase (LDH) release assay over a period of up to 72 hours. Reactive oxygen species formation and antioxidant activity were evaluated using biochemical assays. Metabolomic profiling was performed using proton nuclear magnetic resonance (1H NMR). Results Placental explants exposed to 100 μg/mL of PS-MPs showed a significant increase in cytotoxicity over time (p < 0.01). Levels of mitochondrial and total superoxide anion (p < 0.01 and p < 0.05, respectively) and hydrogen peroxide (p < 0.001) were significantly elevated. PS-MP exposure resulted in a reduction in total sulfhydryl content (p < 0.05) and the activities of antioxidant enzymes superoxide dismutase (p < 0.01) and catalase (p < 0.05), while glutathione peroxidase activity increased (p < 0.05), and the oxidized/reduced glutathione ratio decreased (p < 0.05). Markers of oxidative damage, such as malondialdehyde and carbonylated proteins, also increased significantly (p < 0.001 and p < 0.01, respectively), confirming oxidative stress. Metabolomic analysis revealed significant differences between control and PS-MP-exposed groups, with reduced levels of alanine, formate, glutaric acid, and maltotriose after PS-MP exposure. Discussion This study demonstrates that high concentrations of PS-MPs induce time-dependent cytotoxicity, oxidative stress, and alterations in the TCA cycle, as well as in folate, amino acid, and energy metabolism. These findings highlight the need for further research to clarify the full impact of MP contamination on pregnancy and its implications for future generations.
Collapse
Affiliation(s)
| | - Keyla Silva Nobre Pires
- Cell Biology Laboratory, Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Brazil
| | - Isadora Hart Cavalcante
- Cell Biology Laboratory, Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Brazil
| | | | - Julia Domingues Santos
- Cell Biology Laboratory, Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Brazil
| | | | - Ana Catarina Rezende Leite
- Laboratory of Bioenergetics, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceio, Brazil
| | - Alessandre Carmo Crispim
- Nucleus of Analysis and Research in Nuclear Magnetic Resonance - NAPRMN, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceio, Brazil
| | - Edmilson Rodrigues da Rocha Junior
- Nucleus of Analysis and Research in Nuclear Magnetic Resonance - NAPRMN, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceio, Brazil
| | - Thiago Mendonça Aquino
- Nucleus of Analysis and Research in Nuclear Magnetic Resonance - NAPRMN, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceio, Brazil
| | - Rodrigo Barbano Weingrill
- Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Johann Urschitz
- Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Stephanie Ospina-Prieto
- Cell Biology Laboratory, Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Brazil
| | - Alexandre Urban Borbely
- Cell Biology Laboratory, Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Brazil
| |
Collapse
|
6
|
Salvi S, Fruci S, Lacconi V, Totaro Aprile F, Rullo R, Stuhlmann H, Lanzone A, Campagnolo L, Massimiani M. Effect of Pravastatin on Placental Expression of Epidermal Growth Factor-like Domain 7 in Early-Onset Pre-Eclampsia: A New Potential Mechanism of Action. Biomedicines 2024; 12:1929. [PMID: 39200393 PMCID: PMC11351877 DOI: 10.3390/biomedicines12081929] [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: 07/31/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/02/2024] Open
Abstract
The primary intervention for pre-eclampsia (PE) remains iatrogenic delivery, which can be very preterm and not optimal for the fetus. Although many efforts have been made to prevent and manage PE, there is still a dearth of drugs to treat its pathophysiological progression. Pravastatin (PRA), a hydrophilic statin, has gained interest for the prevention and treatment of PE. The aim of the present study was to evaluate the ability of PRA to modulate factors involved in placentation, such as Epidermal Growth Factor-Like Domain 7 (EGFL7), in human chorionic villous culture from healthy controls and women with PE. A total of 18 women were enrolled: 10 controls and 8 cases. Chorionic villous explants were maintained in culture for 24 h with or without 10 μM Pravastatin, and the expression of EGFL7 and NOTCH1 pathway members was evaluated by qRT-PCR and Western blot analysis. The rationale of the present study was to establish an ex vivo model to identify potential different responses to PRA treatment of chorionic villous explants in order to clarify the molecular mechanism of PRA in the prevention and treatment of PE and to predict whether there are specific clinical conditions that modulate the response to the drug treatment. Within PE patients, two different groups were identified: the high responders, whose villous cultures exhibit significantly increased expressions of the EGFL7 and Notch pathways after PRA incubation; and the low responders, who are high-risk PE patients in which prophylaxis failed to prevent PE and PRA was not able to modulate EGFL7 expression. In conclusion, we identified EGFL7 as a new factor regulated by PRA, placing interest in early discrimination between low- and high- risk women, in which the well-known pharmacological prophylaxis seems to be ineffective, and to explore new potential prevention strategies.
Collapse
Affiliation(s)
- Silvia Salvi
- UOC di Ostetricia e Patologia Ostetrica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, L.go Agostino Gemelli 8, 00168 Rome, Italy; (S.S.); (S.F.); (R.R.); (A.L.)
- Dipartimento Universitario di Scienze della Vita e Sanità Pubblica, Sezione di Ginecologia ed Ostetricia, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy;
| | - Stefano Fruci
- UOC di Ostetricia e Patologia Ostetrica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, L.go Agostino Gemelli 8, 00168 Rome, Italy; (S.S.); (S.F.); (R.R.); (A.L.)
| | - Valentina Lacconi
- Departmental Faculty of Medicine, Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy;
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Federica Totaro Aprile
- Dipartimento Universitario di Scienze della Vita e Sanità Pubblica, Sezione di Ginecologia ed Ostetricia, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy;
| | - Roberta Rullo
- UOC di Ostetricia e Patologia Ostetrica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, L.go Agostino Gemelli 8, 00168 Rome, Italy; (S.S.); (S.F.); (R.R.); (A.L.)
| | - Heidi Stuhlmann
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, Box 60, New York, NY 10065, USA;
| | - Antonio Lanzone
- UOC di Ostetricia e Patologia Ostetrica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, L.go Agostino Gemelli 8, 00168 Rome, Italy; (S.S.); (S.F.); (R.R.); (A.L.)
- Dipartimento Universitario di Scienze della Vita e Sanità Pubblica, Sezione di Ginecologia ed Ostetricia, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy;
| | - Luisa Campagnolo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Micol Massimiani
- Departmental Faculty of Medicine, Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy;
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
7
|
Sun L, Shi M, Wang J, Han X, Wei J, Huang Z, Yang X, Ding Y, Zhang P, He A, Liu M, Yan R, Yang X, Li R, Wang G. Overexpressed Trophoblast Glycoprotein Contributes to Preeclampsia Development by Inducing Abnormal Trophoblast Migration and Invasion Toward the Uterine Spiral Artery. Hypertension 2024; 81:1524-1536. [PMID: 38716674 DOI: 10.1161/hypertensionaha.124.22923] [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: 02/21/2024] [Accepted: 04/22/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Preeclampsia is a significant pregnancy disorder with an unknown cause, mainly attributed to impaired spiral arterial remodeling. METHODS Using RNA sequencing, we identified key genes in placental tissues from healthy individuals and preeclampsia patients. Placenta and plasma samples from pregnant women were collected to detect the expression of TPBG (trophoblast glycoprotein). Pregnant rats were injected with TPBG-carrying adenovirus to detect preeclamptic features. HTR-8/SVneo cells transfected with a TPBG overexpression lentiviral vector were used in cell function experiments. The downstream molecular mechanisms of TPBG were explored using RNA sequencing and single-cell RNA sequencing data. TPBG expression was knocked down in the lipopolysaccharide-induced preeclampsia-like rat model to rescue the preeclampsia features. We also assessed TPBG's potential as an early preeclampsia predictor using clinical plasma samples. RESULTS TPBG emerged as a crucial differentially expressed gene, expressed specifically in syncytiotrophoblasts and extravillous trophoblasts. Subsequently, we established a rat model with preeclampsia-like phenotypes by intravenously injecting TPBG-expressing adenoviruses, observing impaired spiral arterial remodeling, thus indicating a causal correlation between TPBG overexpression and preeclampsia. Studies with HTR-8/SVneo cells, chorionic villous explants, and transwell assays showed TPBG overexpression disrupts trophoblast/extravillous trophoblast migration/invasion and chemotaxis. Notably, TPBG knockdown alleviated the lipopolysaccharide-induced preeclampsia-like rat model. We enhanced preeclampsia risk prediction in early gestation by combining TPBG expression with established clinical predictors. CONCLUSIONS These findings are the first to show that TPBG overexpression contributes to preeclampsia development by affecting uterine spiral artery remodeling. We propose TPBG levels in maternal blood as a predictor of preeclampsia risk. The proposed mechanism by which TPBG overexpression contributes to the occurrence of preeclampsia via its disruptive effect on trophoblast and extravillous trophoblast migration/invasion on uterine spiral artery remodeling, thereby increasing the risk of preeclampsia.
Collapse
Affiliation(s)
- Lu Sun
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Meiting Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Jian Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Xiaoxue Han
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Jiachun Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Zhengrui Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Xiaofeng Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine (P.Z., X.Y., G.W.), Jinan University, Guangzhou, China
| | - Yuzhen Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Ping Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine (P.Z., X.Y., G.W.), Jinan University, Guangzhou, China
| | - Andong He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Mengyuan Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Ruiling Yan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Xuesong Yang
- Key Laboratory for Regenerative Medicine of the Ministry of Education (X.Y., G.W.), Jinan University, Guangzhou, China
- Clinical Research Center, Clifford Hospital, Guangzhou, China (X.Y.)
| | - Ruiman Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine (P.Z., X.Y., G.W.), Jinan University, Guangzhou, China
- Key Laboratory for Regenerative Medicine of the Ministry of Education (X.Y., G.W.), Jinan University, Guangzhou, China
- Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, School of Medicine (G.W.), Jinan University, Guangzhou, China
| |
Collapse
|
8
|
Dugershaw‐Kurzer B, Bossart J, Buljan M, Hannig Y, Zehnder S, Gupta G, Kissling VM, Nowak‐Sliwinska P, van Beijnum JR, Griffioen AW, Masjosthusmann S, Zühr E, Fritsche E, Hornung R, Rduch T, Buerki‐Thurnherr T. Nanoparticles Dysregulate the Human Placental Secretome with Consequences on Angiogenesis and Vascularization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401060. [PMID: 38767187 PMCID: PMC11267331 DOI: 10.1002/advs.202401060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/05/2024] [Indexed: 05/22/2024]
Abstract
Exposure to nanoparticles (NPs) in pregnancy is increasingly linked to adverse effects on embryo-fetal development and health later in life. However, the developmental toxicity mechanisms of NPs are largely unknown, in particular potential effects on the placental secretome, which orchestrates many developmental processes pivotal for pregnancy success. This study demonstrates extensive material- and pregnancy stage-specific deregulation of placental signaling from a single exposure of human placental explants to physiologically relevant concentrations of engineered (silica (SiO2) and titanium dioxide (TiO2) NPs) and environmental NPs (diesel exhaust particles, DEPs). This includes a multitude of secreted inflammatory, vascular, and endocrine placental factors as well as extracellular vesicle (EV)-associated proteins. Moreover, conditioned media (CM) from NP-exposed explants induce pronounced anti-angiogenic and anti-vasculogenic effects, while early neurodevelopmental processes are only marginally affected. These findings underscore the potential of metal oxide NPs and DEPs for widespread interference with the placental secretome and identify vascular morphogenesis as a sensitive outcome for the indirect developmental toxicity of different NPs. Overall, this work has profound implications for the future safety assessment of NPs for industrial, commercial, or medical applications in pregnancy, which should consider placenta-mediated toxicity by holistic secretomics approaches to ensure the development of safe nanotechnologies.
Collapse
Affiliation(s)
- Battuja Dugershaw‐Kurzer
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
- Department of Health Sciences and TechnologyETH ZurichZurich8093Switzerland
| | - Jonas Bossart
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
- Department of Health Sciences and TechnologyETH ZurichZurich8093Switzerland
- SIBSwiss Institute of BioinformaticsLausanne1015Switzerland
| | - Marija Buljan
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
- SIBSwiss Institute of BioinformaticsLausanne1015Switzerland
| | - Yvette Hannig
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
| | - Sarah Zehnder
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
| | - Govind Gupta
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
| | - Vera M. Kissling
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
| | - Patrycja Nowak‐Sliwinska
- Institute of Pharmaceutical Sciences of Western SwitzerlandGeneva1211Switzerland
- School of Pharmaceutical SciencesUniversity of GenevaGeneva1205Switzerland
| | - Judy R. van Beijnum
- Angiogenesis LaboratoryDepartment of Medical OncologyUMC loacation Vrije Universiteit AmsterdamAmsterdam1081The Netherlands
| | - Arjan W. Griffioen
- Angiogenesis LaboratoryDepartment of Medical OncologyUMC loacation Vrije Universiteit AmsterdamAmsterdam1081The Netherlands
| | | | - Etta Zühr
- IUF—Leibniz Research Institute for Environmental Medicine40225DuesseldorfGermany
| | - Ellen Fritsche
- IUF—Leibniz Research Institute for Environmental Medicine40225DuesseldorfGermany
- Medical FacultyHeinrich Heine University40225DuesseldorfGermany
- DNTOX GmbH40223DuesseldorfGermany
| | - René Hornung
- Department of Gynaecology and ObstetricsCantonal Hospital St.Gallen (KSSG)St. Gallen9007Switzerland
| | - Thomas Rduch
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
- Department of Gynaecology and ObstetricsCantonal Hospital St.Gallen (KSSG)St. Gallen9007Switzerland
| | - Tina Buerki‐Thurnherr
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
| |
Collapse
|
9
|
Lapehn S, Nair S, Firsick EJ, MacDonald J, Thoreson C, Litch JA, Bush NR, Kadam L, Girard S, Myatt L, Prasad B, Sathyanarayana S, Paquette AG. Transcriptomic comparison of in vitro models of the human placenta. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.598695. [PMID: 38915703 PMCID: PMC11195179 DOI: 10.1101/2024.06.14.598695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Studying the human placenta through in vitro cell culture methods is necessary due to limited access and amenability of human placental tissue to certain experimental methods as well as distinct anatomical and physiological differences between animal and human placentas. Selecting an in vitro culture model of the human placenta is challenging due to representation of different trophoblast cell types with distinct biological roles and limited comparative studies that define key characteristics of these models. Therefore, the aim of this research was to create a comprehensive transcriptomic comparison of common in vitro models of the human placenta compared to bulk placental tissue from the CANDLE and GAPPS cohorts (N=1083). We performed differential gene expression analysis on publicly available RNA sequencing data from 6 common in vitro models of the human placenta (HTR-8/SVneo, BeWo, JEG-3, JAR, Primary Trophoblasts, and Villous Explants) and compared to CANDLE and GAPPS bulk placental tissue or cytotrophoblast, syncytiotrophoblast, and extravillous trophoblast cell types derived from bulk placental tissue. All in vitro placental models had a substantial number of differentially expressed genes (DEGs, FDR<0.01) compared to the CANDLE and GAPPS placentas (Average DEGs=10,873), and the individual trophoblast cell types (Average DEGs=5,346), indicating that there are vast differences in gene expression compared to bulk and cell-type specific human placental tissue. Hierarchical clustering identified 53 gene clusters with distinct expression profiles across placental models, with 22 clusters enriched for specific KEGG pathways, 7 clusters enriched for high-expression placental genes, and 7 clusters enriched for absorption, distribution, metabolism, and excretion genes. In vitro placental models were classified by fetal sex based on expression of Y-chromosome genes that identified HTR-8/SVneo cells as being of female origin, while JEG-3, JAR, and BeWo cells are of male origin. Overall, none of the models were a close approximation of the transcriptome of bulk human placental tissue, highlighting the challenges with model selection. To enable researchers to select appropriate models, we have compiled data on differential gene expression, clustering, and fetal sex into an accessible web application: "Comparative Transcriptomic Placental Model Atlas (CTPMA)" which can be utilized by researchers to make informed decisions about their selection of in vitro placental models.
Collapse
Affiliation(s)
- Samantha Lapehn
- Center for Developmental Biology and Regenerative Medicine, Seattle Children!s Research Institute, Seattle, WA 98101 United States
| | - Sidharth Nair
- Center for Developmental Biology and Regenerative Medicine, Seattle Children!s Research Institute, Seattle, WA 98101 United States
| | - Evan J. Firsick
- Center for Developmental Biology and Regenerative Medicine, Seattle Children!s Research Institute, Seattle, WA 98101 United States
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA 98195 United States
| | - Ciara Thoreson
- Global Alliance to Prevent Prematurity and Stillbirth, Lynwood, WA 98036 United States
| | - James A Litch
- Global Alliance to Prevent Prematurity and Stillbirth, Lynwood, WA 98036 United States
| | - Nicole R. Bush
- Department of Psychiatry and Behavioral Sciences; Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143 United States
| | - Leena Kadam
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR 97239 United States
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN 55905 United States
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR 97239 United States
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202 United States
| | - Sheela Sathyanarayana
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195 United States
- Center for Child Health, Behavior and Development, Seattle Children!s Research Institute, Seattle, WA 98101 United States
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA 98101 United States
| | - Alison G. Paquette
- Center for Developmental Biology and Regenerative Medicine, Seattle Children!s Research Institute, Seattle, WA 98101 United States
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA 98195 United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195 United States
| |
Collapse
|
10
|
Portillo R, Abad C, Synova T, Kastner P, Heblik D, Kucera R, Karahoda R, Staud F. Cannabidiol disrupts tryptophan metabolism in the human term placenta. Toxicology 2024; 505:153813. [PMID: 38663822 DOI: 10.1016/j.tox.2024.153813] [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: 02/13/2024] [Revised: 04/08/2024] [Accepted: 04/22/2024] [Indexed: 04/30/2024]
Abstract
The increasing use of cannabis during pregnancy raises concerns about its impact on fetal development. While cannabidiol (CBD) shows therapeutic promise, its effects during pregnancy remain uncertain. We investigated CBD's influence on tryptophan (TRP) metabolism in the human placenta. TRP is an essential amino acid that is metabolized via the serotonin and kynurenine (KYN) pathways, which are critical for fetal neurodevelopment. We used human term villous placental explants, an advanced ex vivo model, to study CBD's impact on key TRP metabolic enzymes. In addition, vesicles isolated from the microvillous membrane (MVM) of the human placenta were used to assess CBD's effect on placental serotonin uptake. Explants were exposed to CBD at therapeutic (0.1, 1, 2.5 μg/ml) and non-therapeutic (20 and 40 μg/ml) concentrations to determine its effects on the gene and protein expression of key enzymes in TRP metabolism and metabolite release. CBD upregulated TRP hydroxylase (TPH) and downregulated monoamine oxidase (MAO-A), resulting in reduced levels of 5-hydroxyindoleacetic acid (HIAA). It also downregulated serotonin transporter expression and inhibited serotonin transport across the MVM by up to 60% while simultaneously enhancing TRP metabolism via the kynurenine pathway by upregulating indoleamine-pyrrole 2,3-dioxygenase (IDO-1). Among kynurenine pathway enzymes, kynurenine 3 monooxygenase (KMO) was upregulated while kynurenine aminotransferase 1 (KAT-1) was downregulated; the former is associated with neurotoxic metabolite production, while the latter is linked to reduced neuroprotective metabolite levels. Overall, these results indicate that CBD modulates TRP catabolism in the human placenta, potentially disrupting the tightly regulated homeostasis of the serotonin and KYN pathways.
Collapse
Affiliation(s)
- Ramon Portillo
- Department of Pharmacology and Toxicology, Charles University, Faculty of Pharmacy in Hradec Kralove, Czech Republic
| | - Cilia Abad
- Department of Pharmacology and Toxicology, Charles University, Faculty of Pharmacy in Hradec Kralove, Czech Republic
| | - Tetiana Synova
- Department of Pharmacology and Toxicology, Charles University, Faculty of Pharmacy in Hradec Kralove, Czech Republic
| | - Petr Kastner
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Charles University, Faculty of Pharmacy in Hradec Kralove, Czech Republic
| | - Daniel Heblik
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Charles University, Faculty of Pharmacy in Hradec Kralove, Czech Republic
| | - Radim Kucera
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Charles University, Faculty of Pharmacy in Hradec Kralove, Czech Republic
| | - Rona Karahoda
- Department of Pharmacology and Toxicology, Charles University, Faculty of Pharmacy in Hradec Kralove, Czech Republic
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Charles University, Faculty of Pharmacy in Hradec Kralove, Czech Republic.
| |
Collapse
|
11
|
López-Guzmán C, García AM, Ramirez JD, Aliaga TT, Fernández-Moya A, Kemmerling U, Vásquez AM. Plasmodium falciparum alters the trophoblastic barrier and stroma villi organization of human placental villi explants. Malar J 2024; 23:130. [PMID: 38693572 PMCID: PMC11064279 DOI: 10.1186/s12936-024-04960-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND The sequestration of Plasmodium falciparum infected erythrocytes in the placenta, and the resulting inflammatory response affects maternal and child health. Despite existing information, little is known about the direct impact of P. falciparum on the placental barrier formed by trophoblast and villous stroma. This study aimed to assess placental tissue damage caused by P. falciparum in human placental explants (HPEs). METHODS HPEs from chorionic villi obtained of human term placentas (n = 9) from normal pregnancies were exposed to P. falciparum-infected erythrocytes (IE) for 24 h. HPEs were embedded in paraffin blocks and used to study tissue damage through histopathological and histochemical analysis and apoptosis using TUNEL staining. Culture supernatants were collected to measure cytokine and angiogenic factors and to determine LDH activity as a marker of cytotoxicity. A subset of archived human term placenta paraffin-embedded blocks from pregnant women with malaria were used to confirm ex vivo findings. RESULTS Plasmodium falciparum-IE significantly damages the trophoblast layer and the villous stroma of the chorionic villi. The increased LDH activity and pathological findings such as syncytial knots, fibrin deposits, infarction, trophoblast detachment, and collagen disorganization supported these findings. The specific damage to the trophoblast and the thickening of the subjacent basal lamina were more pronounced in the ex vivo infection. In contrast, apoptosis was higher in the in vivo infection. This disparity could be attributed to the duration of exposure to the infection, which significantly varied between individuals naturally exposed over time and the 24-h exposure in the ex vivo HPE model. CONCLUSION Exposure to P. falciparum-IE induces a detachment of the syncytiotrophoblast, disorganization of the stroma villi, and an increase in apoptosis, alterations that may be associated with adverse results such as intrauterine growth restriction and low birth weight.
Collapse
Affiliation(s)
- Carolina López-Guzmán
- Grupo Malaria, Facultad de Medicina, Universidad de Antioquia, Calle 62 #52-59 Torre 1, Laboratorio 610, Medellín, Colombia
| | - Ana María García
- Grupo Malaria, Facultad de Medicina, Universidad de Antioquia, Calle 62 #52-59 Torre 1, Laboratorio 610, Medellín, Colombia
| | - Juan Diego Ramirez
- Grupo Malaria, Facultad de Medicina, Universidad de Antioquia, Calle 62 #52-59 Torre 1, Laboratorio 610, Medellín, Colombia
| | - Trinidad Torres Aliaga
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile
| | - Alejandro Fernández-Moya
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile
| | - Ulrike Kemmerling
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile
| | - Ana María Vásquez
- Grupo Malaria, Facultad de Medicina, Universidad de Antioquia, Calle 62 #52-59 Torre 1, Laboratorio 610, Medellín, Colombia.
- Escuela de Microbiología, Universidad de Antioquia, Calle 67 # 53-108, Bloque 5, Oficina 5-135, Medellín, Colombia.
| |
Collapse
|
12
|
Kotomura N, Shimono Y, Ishihara S. CYP19A1 Expression Is Controlled by mRNA Stability of the Upstream Transcription Factor AP-2γ in Placental JEG3 Cells. Endocrinology 2024; 165:bqae055. [PMID: 38717933 DOI: 10.1210/endocr/bqae055] [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: 02/14/2024] [Indexed: 05/21/2024]
Abstract
CYP19A1 encodes aromatase, which converts testosterone to estrogen, and is induced during placental maturation. To elucidate the molecular mechanism underlying this function, histone methylation was analyzed using the placental cytotrophoblast cell line, JEG3. Treatment of JEG3 cells with 3-deazaneplanocin A, an inhibitor of several methyltransferases, resulted in increased CYP19A1 expression, accompanied by removal of the repressive mark H3K27me3 from the CYP19A1 promoter. However, this increase was not observed in cells treated with GSK126, another specific inhibitor for H3K27me3 methylation. Expression of TFAP2C, which encodes AP-2γ, a transcription factor that regulates CYP19A1, was also elevated on 3-deazaneplanocin A treatment. Interestingly, TFAP2C messenger RNA (mRNA) was readily degraded in JEG3 cells but protected from degradation in the presence of 3-deazaneplanocin A. TFAP2C mRNA contained N6-methyladenosines, which were reduced on drug treatment. These observations indicate that the TFAP2C mRNA undergoes adenosine methylation and rapid degradation, whereas 3-deazaneplanocin A suppresses methylation, resulting in an increase in AP-2γ levels. We conclude that the increase in AP-2γ expression via stabilization of the TFAP2C mRNA is likely to underlie the increased CYP19A1 expression.
Collapse
Affiliation(s)
- Naoe Kotomura
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake, Aichi 470-1192, Japan
| | - Yohei Shimono
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake, Aichi 470-1192, Japan
| | - Satoru Ishihara
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake, Aichi 470-1192, Japan
| |
Collapse
|
13
|
Andrieu J, Valade M, Lebideau M, Bretelle F, Mège JL, Wurtz N, Mezouar S, La Scola B, Baudoin JP. Pan-microscopic examination of monkeypox virus in trophoblasts cells reveals new insights into virions release through filopodia-like projections. J Med Virol 2024; 96:e29620. [PMID: 38647027 DOI: 10.1002/jmv.29620] [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: 12/19/2023] [Revised: 03/04/2024] [Accepted: 03/31/2024] [Indexed: 04/25/2024]
Abstract
Vertical transmission has been described following monkeypox virus (MPXV) infection in pregnant women. The presence of MPXV has been reported in the placenta from infected women, but whether pathogens colonize placenta remains unexplored. We identify trophoblasts as a target cell for MPXV replication. In a pan-microscopy approach, we decipher the specific infectious cycle of MPXV and inner cellular structures in trophoblasts. We identified the formation of a specialized region for viral morphogenesis and replication in placental cells. We also reported infection-induced cellular remodeling. We found that MPXV stimulates cytoskeleton reorganization with intercellular extensions for MPXV cell spreading specifically to trophoblastic cells. Altogether, the specific infectious cycle of MPXV in trophoblast cells and these protrusions that were structurally and morphologically similar to filopodia reveal new insights into the infection of MPXV.
Collapse
Affiliation(s)
- Jonatane Andrieu
- Centre National de la Recherche Scientifique, Établissement Français du Sang, Anthropologie bio-culturelle, Droit, Éthique et Santé, Aix-Marseille University, Marseille, France
| | - Margaux Valade
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny Infection, Aix-Marseille University, Marseille, France
| | - Marion Lebideau
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny Infection, Aix-Marseille University, Marseille, France
| | - Florence Bretelle
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny Infection, Aix-Marseille University, Marseille, France
- Département de gynécologie et d'obstétrique, Gynépole, La Conception, AP-HM, Marseille, France
| | - Jean-Louis Mège
- Centre National de la Recherche Scientifique, Établissement Français du Sang, Anthropologie bio-culturelle, Droit, Éthique et Santé, Aix-Marseille University, Marseille, France
- Laboratoire d'Immunologie, Assistance Publique Hôpitaux de Marseille (APHM), Marseille, France
| | - Nathalie Wurtz
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny Infection, Aix-Marseille University, Marseille, France
| | - Soraya Mezouar
- Centre National de la Recherche Scientifique, Établissement Français du Sang, Anthropologie bio-culturelle, Droit, Éthique et Santé, Aix-Marseille University, Marseille, France
| | - Bernard La Scola
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny Infection, Aix-Marseille University, Marseille, France
| | - Jean-Pierre Baudoin
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny Infection, Aix-Marseille University, Marseille, France
| |
Collapse
|
14
|
López-Guzmán C, García AM, Marín P, Vásquez AM. Assessment of the Integrity and Function of Human Term Placental Explants in Short-Term Culture. Methods Protoc 2024; 7:16. [PMID: 38392690 PMCID: PMC10892929 DOI: 10.3390/mps7010016] [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: 09/29/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 02/24/2024] Open
Abstract
Human placental explants (HPEs) culture has generated significant interest as a valuable in vitro model for studying tissue functions in response to adverse conditions, such as fluctuations in oxygen levels, nutrient availability, exposure to pathogenic microorganisms, and toxic compounds. HPEs offers the advantage of replicating the intricate microenvironment and cell-to-cell communication involved in this critical and transient organ. Although HPEs culture conditions have been extensively discussed, a protocol for assessing the viability and function of HPEs during short-term culture has not been previously outlined. In this study, we have developed a short-term HPEs culture protocol, specifically up to 72 h, and have employed quantitative, semi-quantitative, and qualitative analyses to evaluate tissue viability and function over time. Under our standardized conditions, placental villi explants began to regain their structural properties (the integrity of the trophoblast and villous stroma) and the functionality of the HPEs (production of angiogenic, endocrine, and immunological factors) starting from 48 h of culture. This restoration ensures a suitable environment for several applications. The data presented here can be highly valuable for laboratories aiming to implement an HPEs model, whether in the process of standardization or seeking to enhance and optimize working conditions and timing with placental tissue.
Collapse
Affiliation(s)
- Carolina López-Guzmán
- Grupo Malaria, Calle 62 # 52-59 Torre 1 Laboratorio 610, Facultad de Medicina, Universidad de Antioquia, Medellín 050001, Colombia; (C.L.-G.); (A.M.G.); (P.M.)
| | - Ana María García
- Grupo Malaria, Calle 62 # 52-59 Torre 1 Laboratorio 610, Facultad de Medicina, Universidad de Antioquia, Medellín 050001, Colombia; (C.L.-G.); (A.M.G.); (P.M.)
| | - Paula Marín
- Grupo Malaria, Calle 62 # 52-59 Torre 1 Laboratorio 610, Facultad de Medicina, Universidad de Antioquia, Medellín 050001, Colombia; (C.L.-G.); (A.M.G.); (P.M.)
| | - Ana María Vásquez
- Grupo Malaria, Calle 62 # 52-59 Torre 1 Laboratorio 610, Facultad de Medicina, Universidad de Antioquia, Medellín 050001, Colombia; (C.L.-G.); (A.M.G.); (P.M.)
- Escuela de Microbiologia, Calle 67 # 53-108, Bloque 5, Oficina 5-135, Universidad de Antioquia, Medellín 050001, Colombia
| |
Collapse
|
15
|
Horcajo P, Ortega-Mora LM, Benavides J, Sánchez-Sánchez R, Amieva R, Collantes-Fernández E, Pastor-Fernández I. Ovine placental explants: A new ex vivo model to study host‒pathogen interactions in reproductive pathogens. Theriogenology 2023; 212:157-171. [PMID: 37729817 DOI: 10.1016/j.theriogenology.2023.09.009] [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: 06/21/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/22/2023]
Abstract
Reproductive failure is one of the main performance constraints in ruminant livestock. Transmissible agents such as Toxoplasma gondii and Neospora caninum are commonly involved in the occurrence of abortion in ruminants, but little is known about the mechanisms involved. While in vivo models are optimal for the study of abortion pathogenesis, they have a high economic cost and come with ethical concerns. Unfortunately, alternative in vitro models fail to replicate the complex in vivo placental structure. To overcome the limitations of currently available models, we developed an ex vivo model based on the cultivation of fresh and cryopreserved sheep placental explants, enabling the biobanking of tissues. Reproducible and simple markers of tissue integrity (histology, RNA concentrations), viability (resazurin reduction), and functionality (synthesis of steroid hormones) were also investigated, allowing a clear quality assessment of the model. This work shows that, similar to fresh explants, tissues cryopreserved in ethylene glycol using slow freezing rates maintain not only their structure and function but also their receptivity to T. gondii and N. caninum infection. In addition, the findings demonstrate that explant lifespan is mainly limited by the culture method, with protocols requiring improvements to extend it beyond 2 days. These findings suggest that cryopreserved tissues can be exploited to study the initial host‒pathogen interactions taking place in the placenta, thus deepening the knowledge of the specific mechanisms that trigger reproductive failure in sheep. Importantly, this work paves the way for the development of similar models in related species and contributes to the reduction of experimental animal use in the future.
Collapse
Affiliation(s)
- Pilar Horcajo
- SALUVET group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Spain.
| | - Luis Miguel Ortega-Mora
- SALUVET group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Spain.
| | - Julio Benavides
- Instituto de Ganadería de Montaña (CSIC-ULE), Grulleros, León, 24346, Spain.
| | - Roberto Sánchez-Sánchez
- SALUVET group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Spain.
| | - Rafael Amieva
- SALUVET group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Spain.
| | - Esther Collantes-Fernández
- SALUVET group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Spain.
| | - Iván Pastor-Fernández
- SALUVET group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Spain.
| |
Collapse
|
16
|
Liu X, Wang G, Huang H, Lv X, Si Y, Bai L, Wang G, Li Q, Yang W. Exploring maternal-fetal interface with in vitro placental and trophoblastic models. Front Cell Dev Biol 2023; 11:1279227. [PMID: 38033854 PMCID: PMC10682727 DOI: 10.3389/fcell.2023.1279227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
The placenta, being a temporary organ, plays a crucial role in facilitating the exchange of nutrients and gases between the mother and the fetus during pregnancy. Any abnormalities in the development of this vital organ not only lead to various pregnancy-related disorders that can result in fetal injury or death, but also have long-term effects on maternal health. In vitro models have been employed to study the physiological features and molecular regulatory mechanisms of placental development, aiming to gain a detailed understanding of the pathogenesis of pregnancy-related diseases. Among these models, trophoblast stem cell culture and organoids show great promise. In this review, we provide a comprehensive overview of the current mature trophoblast stem cell models and emerging organoid models, while also discussing other models in a systematic manner. We believe that this knowledge will be valuable in guiding further exploration of the complex maternal-fetal interface.
Collapse
Affiliation(s)
- Xinlu Liu
- School of Biosciences and Biotechnology, Weifang Medical University, Weifang, Shandong, China
| | - Gang Wang
- Department of Emergency, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Haiqin Huang
- School of Biosciences and Biotechnology, Weifang Medical University, Weifang, Shandong, China
| | - Xin Lv
- School of Biosciences and Biotechnology, Weifang Medical University, Weifang, Shandong, China
| | - Yanru Si
- School of Biosciences and Biotechnology, Weifang Medical University, Weifang, Shandong, China
| | - Lixia Bai
- School of Biosciences and Biotechnology, Weifang Medical University, Weifang, Shandong, China
| | - Guohui Wang
- School of Biosciences and Biotechnology, Weifang Medical University, Weifang, Shandong, China
| | - Qinghua Li
- School of Public Health, Weifang Medical University, Weifang, Shandong, China
| | - Weiwei Yang
- School of Biosciences and Biotechnology, Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
17
|
Tekkatte C, Lindsay SA, Duggan E, Castro-Martínez A, Hakim A, Saldana I, Zhang Y, Zhou J, Sebastian R, Liu Y, Pontigon DS, Meads M, Liu TN, Pizzo DP, Nolan J, Parast MM, Laurent LC. Identification of optimal conditions for human placental explant culture and extracellular vesicle release. iScience 2023; 26:108046. [PMID: 37829201 PMCID: PMC10565782 DOI: 10.1016/j.isci.2023.108046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 05/17/2023] [Accepted: 09/21/2023] [Indexed: 10/14/2023] Open
Abstract
Extracellular vesicles (EVs) can mediate intercellular communication, including signaling between the placenta and maternal tissues. Human placental explant culture is a versatile in vitro model system to investigate placental function. We performed systematic studies in different tissue culture media types and oxygen tensions to identify a defined serum-free culture condition that supports high trophoblast viability and metabolism, as well as the release of similar populations of EVs, compared to traditional undefined conditions that contain media additives potentially contaminated with exogenous EVs. We also determined the time frame in which trophoblast viability and functionality remain optimal. Multiplex vesicle flow cytometry with classical EV and placenta-specific markers revealed three separate populations of explant-derived EVs: small CD63+ EVs; large PLAP+ EVs; and CD63-/PLAP- EVs. These culture and analytical approaches will enable in vitro modeling of short-term effects of environmental perturbations associated with pregnancy complications on placental function and EV release.
Collapse
Affiliation(s)
- Chandana Tekkatte
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Scott A. Lindsay
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Erika Duggan
- Scintillon Institute, San Diego, CA 92121, USA
- Cellarcus Biosciences Inc, La Jolla, CA 92037, USA
| | - Anelizze Castro-Martínez
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Abbas Hakim
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Isabella Saldana
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Yan Zhang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jun Zhou
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Rachel Sebastian
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Yukun Liu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
- Department of Obstetrics and Gynecology, SUN Yat-sen Memorial Hospital, SUN Yat-sen University, Guangzhou, Guangdong, China
| | - Devin S. Pontigon
- Department of Pathology, University of California San Diego, La Jolla, CA 92037, USA
| | - Morgan Meads
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
- Department of Pathology, University of California San Diego, La Jolla, CA 92037, USA
| | - Tzu Ning Liu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Donald P. Pizzo
- Department of Pathology, University of California San Diego, La Jolla, CA 92037, USA
| | - John Nolan
- Scintillon Institute, San Diego, CA 92121, USA
- Cellarcus Biosciences Inc, La Jolla, CA 92037, USA
| | - Mana M. Parast
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
- Department of Pathology, University of California San Diego, La Jolla, CA 92037, USA
| | - Louise C. Laurent
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| |
Collapse
|
18
|
Gonzalez VJ, Li L, Buarpung S, Prahl M, Robinson JF, Gaw SL. Minimal mRNA uptake and inflammatory response to COVID-19 mRNA vaccine exposure in human placental explants. iScience 2023; 26:107549. [PMID: 37664582 PMCID: PMC10470080 DOI: 10.1016/j.isci.2023.107549] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/15/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023] Open
Abstract
Despite universal recommendations for COVID-19 mRNA vaccination in pregnancy, uptake has been lower than desired. There have been limited studies of the direct impact of COVID-19 mRNA vaccine exposure in human placental tissue. Using a primary human placental explants model, we investigated the uptake of two common mRNA vaccines (BNT162b2 Pfizer-BioNTech or mRNA-1273 Moderna), and whether exposure altered villous cytokine responses. Explants derived from second or third trimester chorionic villi were incubated with vaccines at supraphysiologic concentrations and analyzed at two time points. We observed minimal uptake of mRNA vaccines in placental explants by in situ hybridization and quantitative RT-PCR. No specific or global cytokine response was elicited by either of the mRNA vaccines in multiplexed immunoassays. Our results suggest that the human placenta does not readily absorb the COVID-19 mRNA vaccines nor generate a significant inflammatory response after exposure.
Collapse
Affiliation(s)
- Veronica J. Gonzalez
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lin Li
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sirirak Buarpung
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Mary Prahl
- Division of Pediatric Infectious Diseases and Global Health, Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Joshua F. Robinson
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Stephanie L. Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
19
|
Huang L, Tu Z, Wei L, Sun W, Wang Y, Bi S, He F, Du L, Chen J, Kzhyshkowska J, Wang H, Chen D, Zhang S. Generating Functional Multicellular Organoids from Human Placenta Villi. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301565. [PMID: 37438660 PMCID: PMC10502861 DOI: 10.1002/advs.202301565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/26/2023] [Indexed: 07/14/2023]
Abstract
The interaction between trophoblasts, stroma cells, and immune cells at the maternal-fetal interface constitutes the functional units of the placenta, which is crucial for successful pregnancy outcomes. However, the investigation of this intricate interplay is restricted due to the absence of efficient experimental models. To address this challenge, a robust, reliable methodology for generating placenta villi organoids (PVOs) from early, late, or diseased pregnancies using air-liquid surface culture is developed. PVOs contain cytotrophoblasts that can self-renew and differentiate directly, along with stromal elements that retain native immune cells. Analysis of scRNA sequencing and WES data reveals that PVOs faithfully recapitulate the cellular components and genetic alterations of the corresponding source tissue. Additionally, PVOs derived from patients with preeclampsia exhibit specific pathological features such as inflammation, antiangiogenic imbalance, and decreased syncytin expression. The PVO-based propagation of primary placenta villi should enable a deeper investigation of placenta development and exploration of the underlying pathogenesis and therapeutics of placenta-originated diseases.
Collapse
Affiliation(s)
- Lijun Huang
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Zhaowei Tu
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Liudan Wei
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Wei Sun
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Yifan Wang
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Shilei Bi
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Fang He
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Lili Du
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Jingsi Chen
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Julia Kzhyshkowska
- Institute of Transfusion Medicine and ImmunologyMedical Faculty MannheimUniversity of Heidelberg68167MannheimGermany
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health ResearchDepartment of Obstetrics and GynecologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamen361005China
| | - Dunjin Chen
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
- Key Laboratory for Major Obstetric Diseases of Guangdong ProvinceGuangzhou510150China
- Guangdong‐Hong Kong‐Macao Greater Bay Area Higher Education Joint Laboratory of Maternal‐Fetal MedicineGuangzhou510150China
- Guangdong Engineering and Technology Research Center of Maternal‐Fetal MedicineGuangzhou510150China
| | - Shuang Zhang
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| |
Collapse
|
20
|
Staud F, Pan X, Karahoda R, Dong X, Kastner P, Horackova H, Vachalova V, Markert UR, Abad C. Characterization of a human placental clearance system to regulate serotonin levels in the fetoplacental unit. Reprod Biol Endocrinol 2023; 21:74. [PMID: 37612712 PMCID: PMC10464227 DOI: 10.1186/s12958-023-01128-z] [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: 06/28/2023] [Accepted: 08/14/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Serotonin (5-HT) is a biogenic monoamine with diverse functions in multiple human organs and tissues. During pregnancy, tightly regulated levels of 5-HT in the fetoplacental unit are critical for proper placental functions, fetal development, and programming. Despite being a non-neuronal organ, the placenta expresses a suite of homeostatic proteins, membrane transporters and metabolizing enzymes, to regulate monoamine levels. We hypothesized that placental 5-HT clearance is important for maintaining 5-HT levels in the fetoplacental unit. We therefore investigated placental 5-HT uptake from the umbilical circulation at physiological and supraphysiological levels as well as placental metabolism of 5-HT to 5-hydroxyindoleacetic acid (5-HIAA) and 5-HIAA efflux from trophoblast cells. METHODS We employed a systematic approach using advanced organ-, tissue-, and cellular-level models of the human placenta to investigate the transport and metabolism of 5-HT in the fetoplacental unit. Human placentas from uncomplicated term pregnancies were used for perfusion studies, culturing explants, and isolating primary trophoblast cells. RESULTS Using the dually perfused placenta, we observed a high and concentration-dependent placental extraction of 5-HT from the fetal circulation. Subsequently, within the placenta, 5-HT was metabolized to 5-hydroxyindoleacetic acid (5-HIAA), which was then unidirectionally excreted to the maternal circulation. In the explant cultures and primary trophoblast cells, we show concentration- and inhibitor-dependent 5-HT uptake and metabolism and subsequent 5-HIAA release into the media. Droplet digital PCR revealed that the dominant gene in all models was MAO-A, supporting the crucial role of 5-HT metabolism in placental 5-HT clearance. CONCLUSIONS Taken together, we present transcriptional and functional evidence that the human placenta has an efficient 5-HT clearance system involving (1) removal of 5-HT from the fetal circulation by OCT3, (2) metabolism to 5-HIAA by MAO-A, and (3) selective 5-HIAA excretion to the maternal circulation via the MRP2 transporter. This synchronized mechanism is critical for regulating 5-HT in the fetoplacental unit; however, it can be compromised by external insults such as antidepressant drugs.
Collapse
Affiliation(s)
- Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic.
| | - Xin Pan
- Placenta-Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Rona Karahoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Xiaojing Dong
- Placenta-Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Petr Kastner
- Department of Pharmaceutical Chemistry and Drug Analysis, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Hana Horackova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Veronika Vachalova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Udo R Markert
- Placenta-Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| | - Cilia Abad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| |
Collapse
|
21
|
Elzinga FA, Khalili B, Touw DJ, Prins JR, Olinga P, Leuvenink HGD, van Goor H, Gordijn SJ, Nagelkerke A, Mian P. Placenta-on-a-Chip as an In Vitro Approach to Evaluate the Physiological and Structural Characteristics of the Human Placental Barrier upon Drug Exposure: A Systematic Review. J Clin Med 2023; 12:4315. [PMID: 37445348 DOI: 10.3390/jcm12134315] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/23/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Quantification of fetal drug exposure remains challenging since sampling from the placenta or fetus during pregnancy is too invasive. Currently existing in vivo (e.g., cord blood sampling) and ex vivo (e.g., placenta perfusion) models have inherent limitations. A placenta-on-a-chip model is a promising alternative. A systematic search was performed in PubMed on 2 February 2023, and Embase on 14 March 2023. Studies were included where placenta-on-a-chip was used to investigate placental physiology, placenta in different obstetric conditions, and/or fetal exposure to maternally administered drugs. Seventeen articles were included that used comparable approaches but different microfluidic devices and/or different cultured maternal and fetal cell lines. Of these studies, four quantified glucose transfer, four studies evaluated drug transport, three studies investigated nanoparticles, one study analyzed bacterial infection and five studies investigated preeclampsia. It was demonstrated that placenta-on-a-chip has the capacity to recapitulate the key characteristics of the human placental barrier. We aimed to identify knowledge gaps and provide the first steps towards an overview of current protocols for developing a placenta-on-a-chip, that facilitates comparison of results from different studies. Although models differ, they offer a promising approach for in vitro human placental and fetal drug studies under healthy and pathological conditions.
Collapse
Affiliation(s)
- Femke A Elzinga
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Behrad Khalili
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Daan J Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deunsinglaan 1, 9713 AV Groningen, The Netherlands
| | - Jelmer R Prins
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deunsinglaan 1, 9713 AV Groningen, The Netherlands
| | - Henri G D Leuvenink
- Department of Surgery, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, Pathology Section, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Sanne J Gordijn
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Anika Nagelkerke
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deunsinglaan 1, 9713 AV Groningen, The Netherlands
| | - Paola Mian
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
22
|
Forstner D, Guettler J, Brugger BA, Lyssy F, Neuper L, Daxboeck C, Cvirn G, Fuchs J, Kraeker K, Frolova A, Valdes DS, Stern C, Hirschmugl B, Fluhr H, Wadsack C, Huppertz B, Nonn O, Herse F, Gauster M. CD39 abrogates platelet-derived factors induced IL-1β expression in the human placenta. Front Cell Dev Biol 2023; 11:1183793. [PMID: 37325567 PMCID: PMC10264854 DOI: 10.3389/fcell.2023.1183793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/16/2023] [Indexed: 06/17/2023] Open
Abstract
Tissue insults in response to inflammation, hypoxia and ischemia are accompanied by the release of ATP into the extracellular space. There, ATP modulates several pathological processes, including chemotaxis, inflammasome induction and platelet activation. ATP hydrolysis is significantly enhanced in human pregnancy, suggesting that increased conversion of extracellular ATP is an important anti-inflammatory process in preventing exaggerated inflammation, platelet activation and hemostasis in gestation. Extracellular ATP is converted into AMP, and subsequently into adenosine by the two major nucleotide-metabolizing enzymes CD39 and CD73. Here, we aimed to elucidate developmental changes of placental CD39 and CD73 over gestation, compared their expression in placental tissue from patients with preeclampsia and healthy controls, and analyzed their regulation in response to platelet-derived factors and different oxygen conditions in placental explants as well as the trophoblast cell line BeWo. Linear regression analysis showed a significant increase in placental CD39 expression, while at the same time CD73 levels declined at term of pregnancy. Neither maternal smoking during first trimester, fetal sex, maternal age, nor maternal BMI revealed any effects on placental CD39 and CD73 expression. Immunohistochemistry detected both, CD39 and CD73, predominantly in the syncytiotrophoblast layer. Placental CD39 and CD73 expression were significantly increased in pregnancies complicated with preeclampsia, when compared to controls. Cultivation of placental explants under different oxygen conditions had no effect on the ectonucleotidases, whereas presence of platelet releasate from pregnant women led to deregulated CD39 expression. Overexpression of recombinant human CD39 in BeWo cells decreased extracellular ATP levels after culture in presence of platelet-derived factors. Moreover, platelet-derived factors-induced upregulation of the pro-inflammatory cytokine, interleukin-1β, was abolished by CD39 overexpression. Our study shows that placental CD39 is upregulated in preeclampsia, suggesting an increasing demand for extracellular ATP hydrolysis at the utero-placental interface. Increased placental CD39 in response to platelet-derived factors may lead to enhanced conversion of extracellular ATP levels, which in turn could represent an important anti-coagulant defense mechanism of the placenta.
Collapse
Affiliation(s)
- Désirée Forstner
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Jacqueline Guettler
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Beatrice A. Brugger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Freya Lyssy
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Lena Neuper
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Christine Daxboeck
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Gerhard Cvirn
- Division of Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Julia Fuchs
- Division of Medical Physics and Biophysics, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Kristin Kraeker
- Experimental and Clinical Research Center, A Cooperation Between the Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association and the Charité—Universitätsmedizin Berlin, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, Berlin, Germany
| | - Alina Frolova
- Experimental and Clinical Research Center, A Cooperation Between the Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association and the Charité—Universitätsmedizin Berlin, Berlin, Germany
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute of Molecular Biology and Genetic of NASU, Kyiv, Ukraine
| | - Daniela S. Valdes
- Experimental and Clinical Research Center, A Cooperation Between the Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association and the Charité—Universitätsmedizin Berlin, Berlin, Germany
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Christina Stern
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Birgit Hirschmugl
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Herbert Fluhr
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Berthold Huppertz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Olivia Nonn
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
- Experimental and Clinical Research Center, A Cooperation Between the Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association and the Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Herse
- Experimental and Clinical Research Center, A Cooperation Between the Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association and the Charité—Universitätsmedizin Berlin, Berlin, Germany
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Martin Gauster
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
23
|
High Glucose Promotes Inflammation and Weakens Placental Defenses against E. coli and S. agalactiae Infection: Protective Role of Insulin and Metformin. Int J Mol Sci 2023; 24:ijms24065243. [PMID: 36982317 PMCID: PMC10048930 DOI: 10.3390/ijms24065243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/22/2023] [Accepted: 02/28/2023] [Indexed: 03/12/2023] Open
Abstract
Placentas from gestational diabetes mellitus (GDM) patients undergo significant metabolic and immunologic adaptations due to hyperglycemia, which results in an exacerbated synthesis of proinflammatory cytokines and an increased risk for infections. Insulin or metformin are clinically indicated for the treatment of GDM; however, there is limited information about the immunomodulatory activity of these drugs in the human placenta, especially in the context of maternal infections. Our objective was to study the role of insulin and metformin in the placental inflammatory response and innate defense against common etiopathological agents of pregnancy bacterial infections, such as E. coli and S. agalactiae, in a hyperglycemic environment. Term placental explants were cultivated with glucose (10 and 50 mM), insulin (50–500 nM) or metformin (125–500 µM) for 48 h, and then they were challenged with live bacteria (1 × 105 CFU/mL). We evaluated the inflammatory cytokine secretion, beta defensins production, bacterial count and bacterial tissue invasiveness after 4–8 h of infection. Our results showed that a GDM-associated hyperglycemic environment induced an inflammatory response and a decreased beta defensins synthesis unable to restrain bacterial infection. Notably, both insulin and metformin exerted anti-inflammatory effects under hyperglycemic infectious and non-infectious scenarios. Moreover, both drugs fortified placental barrier defenses, resulting in reduced E. coli counts, as well as decreased S. agalactiae and E. coli invasiveness of placental villous trees. Remarkably, the double challenge of high glucose and infection provoked a pathogen-specific attenuated placental inflammatory response in the hyperglycemic condition, mainly denoted by reduced TNF-α and IL-6 secretion after S. agalactiae infection and by IL-1β after E. coli infection. Altogether, these results suggest that metabolically uncontrolled GDM mothers develop diverse immune placental alterations, which may help to explain their increased vulnerability to bacterial pathogens.
Collapse
|
24
|
Dusza HM, van Boxel J, van Duursen MBM, Forsberg MM, Legler J, Vähäkangas KH. Experimental human placental models for studying uptake, transport and toxicity of micro- and nanoplastics. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 860:160403. [PMID: 36417947 DOI: 10.1016/j.scitotenv.2022.160403] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 06/16/2023]
Abstract
Micro- and nanoplastics (MNPs) are ubiquitous in the environment and have recently been found in human lungs, blood and placenta. However, data on the possible effects of MNPs on human health is extremely scarce. The potential toxicity of MNPs during pregnancy, a period of increased susceptibility to environmental insults, is of particular concern. The placenta provides a unique interface between maternal and fetal circulation which is essential for in utero survival and healthy pregnancy. Placental toxicokinetics and toxicity of MNPs are still largely unexplored and the limited studies performed up to now focus mainly on polystyrene particles. Practical and ethical considerations limit research options in humans, and extrapolation from animal studies is challenging due to marked differences between species. Nevertheless, diverse in vitro and ex vivo human placental models exist e.g., plasma membrane vesicles, mono-culture and co-culture of placental cells, placenta-on-a-chip, villous tissue explants, and placental perfusion that can be used to advance this research area. The objective of this concise review is to recapitulate different human placental models, summarize the current understanding of placental uptake, transport and toxicity of MNPs and define knowledge gaps. Moreover, we provide perspectives for future research urgently needed to assess the potential hazards and risks of MNP exposure to maternal and fetal health.
Collapse
Affiliation(s)
- Hanna M Dusza
- Division of Toxicology, Institute for Risk Assessment Sciences, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| | - Jeske van Boxel
- Amsterdam Institute for Life and Environment, Faculty of Science, Vrije Universiteit Amsterdam, the Netherlands
| | - Majorie B M van Duursen
- Amsterdam Institute for Life and Environment, Faculty of Science, Vrije Universiteit Amsterdam, the Netherlands
| | - Markus M Forsberg
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Juliette Legler
- Division of Toxicology, Institute for Risk Assessment Sciences, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Kirsi H Vähäkangas
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
25
|
Gonzalez V, Li L, Buarpung S, Prahl M, Robinson JF, Gaw SL. Minimal mRNA uptake and inflammatory response to COVID-19 mRNA vaccine exposure in human placental explants. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.01.23285349. [PMID: 36778281 PMCID: PMC9915836 DOI: 10.1101/2023.02.01.23285349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Despite universal recommendations for COVID-19 mRNA vaccination in pregnancy, uptake has been lower than desired. There have been limited studies of the direct impact of COVID-19 mRNA vaccine exposure in human placental tissue. Using a primary human villous explant model, we investigated the uptake of two common mRNA vaccines (BNT162b2 Pfizer-BioNTech or mRNA-1273 Moderna), and whether exposure altered villous cytokine responses. Explants derived from second or third trimester chorionic villi were incubated with vaccines at supraphysiologic concentrations and analyzed at two time points. We observed minimal uptake of mRNA vaccines in placental explants by in situ hybridization and quantitative RT-PCR. No specific or global cytokine response was elicited by either of the mRNA vaccines in multiplexed immunoassays. Our results suggest that the human placenta does not readily absorb the COVID-19 mRNA vaccines nor generate a significant inflammatory response after exposure.
Collapse
Affiliation(s)
- Veronica Gonzalez
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, California 94158, USA
- These authors contributed equally to this work
| | - Lin Li
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, California 94158, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, California 94143, USA
- These authors contributed equally to this work
| | - Sirirak Buarpung
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, California 94158, USA
| | - Mary Prahl
- Division of Pediatric Infectious Diseases and Global Health, Department of Pediatrics, University of California, San Francisco, California 94110, USA
| | - Joshua F. Robinson
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, California 94143, USA
| | - Stephanie L. Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, California 94158, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, California 94143, USA
| |
Collapse
|
26
|
Cherubini M, Haase K. A Bioengineered Model for Studying Vascular-Pericyte Interactions of the Placenta. Methods Mol Biol 2023; 2608:409-423. [PMID: 36653720 DOI: 10.1007/978-1-0716-2887-4_23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Investigating the complex cellular interactions of the placenta has remained, until now, a challenge in the field. Given the ethical limitations of studying human placentae, and the interspecies differences that exist between mammals, in vitro models are a valuable tool for investigating developmental and pathologic processes related to the human placenta. A number of in vitro models have been recently employed to investigate various aspects of placental development, with many focusing on the maternal-fetal interface including the trophoblasts and an endothelial barrier. One critical aspect in mimicking the physiology of the placenta is to include perfusable microvessels. As this organ is highly vascularized, it is pertinent to represent the exchange of oxygen and nutrients from the maternal blood to the embedded vessels of the fetus. Using hydrogel-laden microfluidics, it is now possible to bioengineer these and other microvessels in a reproducible manner. By using HUVEC, fetal-like vessels can be generated on a chip and can be studied in a controlled manner. This chapter introduces the concept of generating a triculture vasculature on-chip system, which can be employed to study placental pericyte-endothelial interactions. We describe strategies for generating the vessels on-chip, as well as for quantifying vascular morphology and function. This methodology allows for unique microvessel-related biological questions to be addressed, including how stromal cells impact vascular remodeling over time.
Collapse
Affiliation(s)
- Marta Cherubini
- European Molecular Biology Laboratory (EMBL), Barcelona, Spain
| | - Kristina Haase
- European Molecular Biology Laboratory (EMBL), Barcelona, Spain.
| |
Collapse
|
27
|
Krausová M, Braun D, Buerki-Thurnherr T, Gundacker C, Schernhammer E, Wisgrill L, Warth B. Understanding the Chemical Exposome During Fetal Development and Early Childhood: A Review. Annu Rev Pharmacol Toxicol 2023; 63:517-540. [PMID: 36202091 DOI: 10.1146/annurev-pharmtox-051922-113350] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Early human life is considered a critical window of susceptibility to external exposures. Infants are exposed to a multitude of environmental factors, collectively referred to as the exposome. The chemical exposome can be summarized as the sum of all xenobiotics that humans are exposed to throughout a lifetime. We review different exposure classes and routes that impact fetal and infant metabolism and the potential toxicological role of mixture effects. We also discuss the progress in human biomonitoring and present possiblemodels for studying maternal-fetal transfer. Data gaps on prenatal and infant exposure to xenobiotic mixtures are identified and include natural biotoxins, in addition to commonly reported synthetic toxicants, to obtain a more holistic assessment of the chemical exposome. We highlight the lack of large-scale studies covering a broad range of xenobiotics. Several recommendations to advance our understanding of the early-life chemical exposome and the subsequent impact on health outcomes are proposed.
Collapse
Affiliation(s)
- Magdaléna Krausová
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria; , ,
| | - Dominik Braun
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria; , ,
| | - Tina Buerki-Thurnherr
- Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Particles Biology Interactions, St. Gallen, Switzerland;
| | - Claudia Gundacker
- Center for Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria; .,Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria
| | - Eva Schernhammer
- Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria.,Center for Public Health, Department of Epidemiology, Medical University of Vienna, Vienna, Austria; .,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Lukas Wisgrill
- Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria.,Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria;
| | - Benedikt Warth
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria; , , .,Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria
| |
Collapse
|
28
|
Luconi M, Sogorb MA, Markert UR, Benfenati E, May T, Wolbank S, Roncaglioni A, Schmidt A, Straccia M, Tait S. Human-Based New Approach Methodologies in Developmental Toxicity Testing: A Step Ahead from the State of the Art with a Feto-Placental Organ-on-Chip Platform. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:15828. [PMID: 36497907 PMCID: PMC9737555 DOI: 10.3390/ijerph192315828] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/20/2022] [Accepted: 11/25/2022] [Indexed: 06/17/2023]
Abstract
Developmental toxicity testing urgently requires the implementation of human-relevant new approach methodologies (NAMs) that better recapitulate the peculiar nature of human physiology during pregnancy, especially the placenta and the maternal/fetal interface, which represent a key stage for human lifelong health. Fit-for-purpose NAMs for the placental-fetal interface are desirable to improve the biological knowledge of environmental exposure at the molecular level and to reduce the high cost, time and ethical impact of animal studies. This article reviews the state of the art on the available in vitro (placental, fetal and amniotic cell-based systems) and in silico NAMs of human relevance for developmental toxicity testing purposes; in addition, we considered available Adverse Outcome Pathways related to developmental toxicity. The OECD TG 414 for the identification and assessment of deleterious effects of prenatal exposure to chemicals on developing organisms will be discussed to delineate the regulatory context and to better debate what is missing and needed in the context of the Developmental Origins of Health and Disease hypothesis to significantly improve this sector. Starting from this analysis, the development of a novel human feto-placental organ-on-chip platform will be introduced as an innovative future alternative tool for developmental toxicity testing, considering possible implementation and validation strategies to overcome the limitation of the current animal studies and NAMs available in regulatory toxicology and in the biomedical field.
Collapse
Affiliation(s)
- Michaela Luconi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
- I.N.B.B. (Istituto Nazionale Biostrutture e Biosistemi), Viale Medaglie d’Oro 305, 00136 Rome, Italy
| | - Miguel A. Sogorb
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Avenida de la Universidad s/n, 03202 Elche, Spain
| | - Udo R. Markert
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Emilio Benfenati
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Tobias May
- InSCREENeX GmbH, Inhoffenstr. 7, 38124 Braunschweig, Germany
| | - Susanne Wolbank
- Ludwig Boltzmann Institut for Traumatology, The Research Center in Cooperation with AUVA, Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Alessandra Roncaglioni
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Astrid Schmidt
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Marco Straccia
- FRESCI by Science&Strategy SL, C/Roure Monjo 33, Vacarisses, 08233 Barcelona, Spain
| | - Sabrina Tait
- Centre for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| |
Collapse
|
29
|
Renaud SJ, Jeyarajah MJ. How trophoblasts fuse: an in-depth look into placental syncytiotrophoblast formation. Cell Mol Life Sci 2022; 79:433. [PMID: 35859055 PMCID: PMC11072895 DOI: 10.1007/s00018-022-04475-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/07/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022]
Abstract
In humans, cell fusion is restricted to only a few cell types under normal conditions. In the placenta, cell fusion is a critical process for generating syncytiotrophoblast: the giant multinucleated trophoblast lineage containing billions of nuclei within an interconnected cytoplasm that forms the primary interface separating maternal blood from fetal tissue. The unique morphology of syncytiotrophoblast ensures that nutrients and gases can be efficiently transferred between maternal and fetal tissue while simultaneously restricting entry of potentially damaging substances and maternal immune cells through intercellular junctions. To maintain integrity of the syncytiotrophoblast layer, underlying cytotrophoblast progenitor cells terminate their capability for self-renewal, upregulate expression of genes needed for differentiation, and then fuse into the overlying syncytium. These processes are disrupted in a variety of obstetric complications, underscoring the importance of proper syncytiotrophoblast formation for pregnancy health. Herein, an overview of key mechanisms underlying human trophoblast fusion and syncytiotrophoblast development is discussed.
Collapse
Affiliation(s)
- Stephen J Renaud
- Department of Anatomy and Cell Biology and Children's Health Research Institute, University of Western Ontario, London, ON, N6A5C1, Canada.
| | - Mariyan J Jeyarajah
- Department of Anatomy and Cell Biology and Children's Health Research Institute, University of Western Ontario, London, ON, N6A5C1, Canada
| |
Collapse
|
30
|
Gonçalves BM, Graceli JB, da Rocha PB, Tilli HP, Vieira EM, de Sibio MT, Peghinelli VV, Deprá IC, Mathias LS, Olímpio RMC, Belik VC, Nogueira CR. Placental model as an important tool to study maternal-fetal interface. Reprod Toxicol 2022; 112:7-13. [PMID: 35714933 DOI: 10.1016/j.reprotox.2022.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/29/2022] [Accepted: 06/09/2022] [Indexed: 10/18/2022]
Abstract
The placenta is a temporary organ that plays critical roles at the maternal-fetal interface. Normal development and function of the placenta is dependent on hormonal signaling pathways that make the placenta a target of endocrine disrupting chemical (EDC) action. Studies showing association between prenatal exposure, hormone disruption, and reproductive damage indicate that EDCs are developmentally toxic and can impact future generations. In this context, new placental models (trophoblast-derived cell lines, organotypic or 3D cell models, and physiologically based kinetic models) have been developed in order to create new approach methodology (NAM) to assess and even prevent such disastrous toxic harm in future generations. With the widespread discouragement of conducting animal studies, it has become irrefutable to develop in vitro models that can serve as a substitute for in vivo models. The goal of this review is to discuss the newest in vitro models to understand the maternal-fetal interface and predict placental development, physiology, and dysfunction generated by failures in molecular hormone control mechanisms, which, consequently, may change epigenetic programming to increase susceptibility to metabolic and other disorders in the offspring. We summarize the latest placental models for developmental toxicology studies, focusing mainly on three-dimensional (3D) culture models.
Collapse
Affiliation(s)
- Bianca M Gonçalves
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil.
| | - Jones B Graceli
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo (UFES), Vitória, ES, Brazil
| | - Paula B da Rocha
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Helena P Tilli
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Ester M Vieira
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Maria T de Sibio
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Vinícius V Peghinelli
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Igor C Deprá
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Lucas S Mathias
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Regiane M C Olímpio
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Virgínia C Belik
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Célia R Nogueira
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil.
| |
Collapse
|
31
|
Fliedel L, Alhareth K, Mignet N, Fournier T, Andrieux K. Placental Models for Evaluation of Nanocarriers as Drug Delivery Systems for Pregnancy Associated Disorders. Biomedicines 2022; 10:936. [PMID: 35625672 PMCID: PMC9138319 DOI: 10.3390/biomedicines10050936] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/06/2022] [Accepted: 04/18/2022] [Indexed: 12/12/2022] Open
Abstract
Pregnancy-associated disorders affect around 20% of pregnancies each year around the world. The risk associated with pregnancy therapeutic management categorizes pregnant women as "drug orphan" patients. In the last few decades, nanocarriers have demonstrated relevant properties for controlled drug delivery, which have been studied for pregnancy-associated disorders. To develop new drug dosage forms it is mandatory to have access to the right evaluation models to ensure their usage safety and efficacy. This review exposes the various placental-based models suitable for nanocarrier evaluation for pregnancy-associated therapies. We first review the current knowledge about nanocarriers as drug delivery systems and how placenta can be used as an evaluation model. Models are divided into three categories: in vivo, in vitro, and ex vivo placental models. We then examine the recent studies using those models to evaluate nanocarriers behavior towards the placental barrier and which information can be gathered from these results. Finally, we propose a flow chart on the usage and the combination of models regarding the nanocarriers and nanoparticles studied and the intended therapeutic strategy.
Collapse
Affiliation(s)
- Louise Fliedel
- Unité de Technologies Chimiques et Biologiques Pour la Santé (UTCBS), Inserm U1267, CNRS UMR8258, Faculté de Pharmacie, Université de Paris Cité, 75006 Paris, France; (L.F.); (K.A.); (N.M.)
- Pathophysiology and Pharmacotoxicology of the Human Placenta, Pre and Postnatal Microbiota Unit (3PHM), Inserm U1139, Faculté de Pharmacie, Université de Paris Cité, 75006 Paris, France;
| | - Khair Alhareth
- Unité de Technologies Chimiques et Biologiques Pour la Santé (UTCBS), Inserm U1267, CNRS UMR8258, Faculté de Pharmacie, Université de Paris Cité, 75006 Paris, France; (L.F.); (K.A.); (N.M.)
| | - Nathalie Mignet
- Unité de Technologies Chimiques et Biologiques Pour la Santé (UTCBS), Inserm U1267, CNRS UMR8258, Faculté de Pharmacie, Université de Paris Cité, 75006 Paris, France; (L.F.); (K.A.); (N.M.)
| | - Thierry Fournier
- Pathophysiology and Pharmacotoxicology of the Human Placenta, Pre and Postnatal Microbiota Unit (3PHM), Inserm U1139, Faculté de Pharmacie, Université de Paris Cité, 75006 Paris, France;
| | - Karine Andrieux
- Unité de Technologies Chimiques et Biologiques Pour la Santé (UTCBS), Inserm U1267, CNRS UMR8258, Faculté de Pharmacie, Université de Paris Cité, 75006 Paris, France; (L.F.); (K.A.); (N.M.)
| |
Collapse
|
32
|
Bačenková D, Trebuňová M, Čížková D, Hudák R, Dosedla E, Findrik-Balogová A, Živčák J. In Vitro Model of Human Trophoblast in Early Placentation. Biomedicines 2022; 10:biomedicines10040904. [PMID: 35453654 PMCID: PMC9029210 DOI: 10.3390/biomedicines10040904] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/09/2022] [Accepted: 04/13/2022] [Indexed: 12/26/2022] Open
Abstract
The complex process of placental implantation and development affects trophoblast progenitors and uterine cells through the regulation of transcription factors, cytokines, adhesion receptors and their ligands. Differentiation of trophoblast precursors in the trophectoderm of early ontogenesis, caused by the transcription factors, such as CDX2, TEAD4, Eomes and GATA3, leads to the formation of cytotrophoblast and syncytiotrophoblast populations. The molecular mechanisms involved in placental formation inside the human body along with the specification and differentiation of trophoblast cell lines are, mostly due to the lack of suitable cell models, not sufficiently elucidated. This review is an evaluation of current technologies, which are used to study the behavior of human trophoblasts and other placental cells, as well as their ability to represent physiological conditions both in vivo and in vitro. An in vitro 3D model with a characteristic phenotype is of great benefit for the study of placental physiology. At the same time, it provides great support for future modeling of placental disease.
Collapse
Affiliation(s)
- Darina Bačenková
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, 04200 Košice, Slovakia; (M.T.); (R.H.); (A.F.-B.); (J.Ž.)
- Correspondence: ; Tel.: +42-1055-602-2380
| | - Marianna Trebuňová
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, 04200 Košice, Slovakia; (M.T.); (R.H.); (A.F.-B.); (J.Ž.)
| | - Daša Čížková
- Centre for Experimental and Clinical Regenerative Medicine, The University of Veterinary Medicine and Pharmacy, 04181 Košice, Slovakia;
| | - Radovan Hudák
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, 04200 Košice, Slovakia; (M.T.); (R.H.); (A.F.-B.); (J.Ž.)
| | - Erik Dosedla
- Department of Gynecology and Obstetrics, Faculty of Medicine, Pavol Jozef Šafarik Univerzity Hospital AGEL Košice-Šaca, Pavol Jozef Šafarik University in Košice, 04015 Košice-Šaca, Slovakia;
| | - Alena Findrik-Balogová
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, 04200 Košice, Slovakia; (M.T.); (R.H.); (A.F.-B.); (J.Ž.)
| | - Jozef Živčák
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, 04200 Košice, Slovakia; (M.T.); (R.H.); (A.F.-B.); (J.Ž.)
| |
Collapse
|
33
|
Langerak T, Broekhuizen M, Unger PPA, Tan L, Koopmans M, van Gorp E, Danser AHJ, Rockx B. Transplacental Zika virus transmission in ex vivo perfused human placentas. PLoS Negl Trop Dis 2022; 16:e0010359. [PMID: 35442976 PMCID: PMC9060339 DOI: 10.1371/journal.pntd.0010359] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/02/2022] [Accepted: 03/25/2022] [Indexed: 11/18/2022] Open
Abstract
A Zika virus (ZIKV) infection during pregnancy can result in severe birth defects such as microcephaly. To date, it is incompletely understood how ZIKV can cross the human placenta. Furthermore, results from studies in pregnant mice and non-human primates are conflicting regarding the role of cross-reactive dengue virus (DENV) antibodies on transplacental ZIKV transmission. Elucidating how ZIKV can cross the placenta and which risk factors contribute to this is important for risk assessment and for potential intervention strategies for transplacental ZIKV transmission. In this study we use an ex vivo human placental perfusion model to study transplacental ZIKV transmission and the effect that cross-reactive DENV antibodies have on this transmission. By using this model, we demonstrate that DENV antibodies significantly increase ZIKV uptake in perfused human placentas and that this increased uptake is neonatal Fc-receptor-dependent. Furthermore, we show that cross-reactive DENV antibodies enhance ZIKV infection in term human placental explants and in primary fetal macrophages but not in primary trophoblasts. Our data supports the hypothesis that presence of cross-reactive DENV antibodies could be an important risk factor for transplacental ZIKV transmission. Furthermore, we demonstrate that the ex vivo placental perfusion model is a relevant and animal friendly model to study transplacental pathogen transmission. Zika virus is a mosquito-transmitted virus that can cause severe birth defects such as microcephaly when the infection occurs during pregnancy. Understanding how Zika virus crosses the placenta during pregnancy is important for future prevention strategies for vertical Zika virus transmission. Despite significant efforts to study this, to date it remains incompletely understood how Zika virus can cross the placenta and which risk factors contribute to this form of transmission. In this study we use an ex vivo placental perfusion model to study transplacental Zika virus transmission. The ex vivo placental perfusion model is a highly physiological and animal friendly model that mimics the in vivo conditions during pregnancy. We found that antibodies against the closely related dengue virus can significantly enhance placental uptake of Zika virus and Zika virus infection of human placental explants and fetal macrophages. These findings indicate that presence of cross-reactive dengue virus antibodies could contribute to transplacental Zika virus transmission.
Collapse
Affiliation(s)
- Thomas Langerak
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Michelle Broekhuizen
- Department of Internal Medicine, Division of Pharmacology, Erasmus Medical Center, Rotterdam, the Netherlands
- Department of Pediatrics, Division of Neonatology, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Lunbo Tan
- Department of Internal Medicine, Division of Pharmacology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Marion Koopmans
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Eric van Gorp
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - A. H. Jan Danser
- Department of Internal Medicine, Division of Pharmacology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Barry Rockx
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
- * E-mail:
| |
Collapse
|
34
|
Animal Models of Chorioamnionitis: Considerations for Translational Medicine. Biomedicines 2022; 10:biomedicines10040811. [PMID: 35453561 PMCID: PMC9032938 DOI: 10.3390/biomedicines10040811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 02/04/2023] Open
Abstract
Preterm birth is defined as any birth occurring before 37 completed weeks of gestation by the World Health Organization. Preterm birth is responsible for perinatal mortality and long-term neurological morbidity. Acute chorioamnionitis is observed in 70% of premature labor and is associated with a heavy burden of multiorgan morbidities in the offspring. Unfortunately, chorioamnionitis is still missing effective biomarkers and early placento- as well as feto-protective and curative treatments. This review summarizes recent advances in the understanding of the underlying mechanisms of chorioamnionitis and subsequent impacts on the pregnancy outcome, both during and beyond gestation. This review also describes relevant and current animal models of chorioamnionitis used to decipher associated mechanisms and develop much needed therapies. Improved knowledge of the pathophysiological mechanisms underpinning chorioamnionitis based on preclinical models is a mandatory step to identify early in utero diagnostic biomarkers and design novel anti-inflammatory interventions to improve both maternal and fetal outcomes.
Collapse
|
35
|
Liu L, Wang R, Xu R, Chu Y, Gu W. Procyanidin B2 ameliorates endothelial dysfunction and impaired angiogenesis via the Nrf2/PPARγ/sFlt-1 axis in preeclampsia. Pharmacol Res 2022; 177:106127. [PMID: 35150862 DOI: 10.1016/j.phrs.2022.106127] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/30/2022] [Accepted: 02/08/2022] [Indexed: 12/14/2022]
Abstract
Preeclampsia is a severe complication of pregnancy characterized by variable degrees of placental malperfusion. A growing body of evidence indicates that soluble endoglin and soluble fms-like tyrosine kinase-1 (sFlt-1) play important pathophysiological roles in preeclampsia, causing endothelial dysfunction, hypertension, and multiorgan injury. A drug that is safe in pregnancy and inhibits placental sFlt-1 and soluble endoglin secretion would be an attractive treatment strategy for preeclampsia. Procyanidin B2, a bioactive food compound, has been reported to exert multiple beneficial functions. Placental explant cultures in vitro are useful for studying tissue functions including release of secretory components, pharmacology, toxicology, and disease processes. The reduced uterine perfusion pressure (RUPP) rat model has been widely used as a model of preeclampsia. We aimed to investigate the effect of procyanidin B2 on preeclampsia via using placental explant cultures and RUPP rat model. In this study, we demonstrated that procyanidin B2 reduced soluble endoglin and sFlt-1 secretion from human umbilical vein endothelial cells (HUVECs), primary trophoblasts, and placental explants from preeclamptic pregnancies. Moreover, procyanidin B2 alleviated endothelial dysfunction and impaired angiogenesis induced by sFlt-1, including increasing the migration, invasion and angiogenesis of endothelial cells and decreasing the expression of vascular cell adhesion molecule-1 (VCAM-1) and leukocyte adhesion on HUVECs. In addition, procyanidin B2 promoted nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear accumulation and induced peroxisome proliferator-activated receptor γ (PPARγ) expression in primary placental tissues and endothelial cells. Importantly, Nrf2 specifically binds to the PPARγ promoter region (-1227/-1217) and enhances its transcriptional activity. Procyanidin B2 inhibits sFlt-1 secretion via the Nrf2/PPARγ axis. In the RUPP rat model of preeclampsia, procyanidin B2 attenuated RUPP-induced maternal angiogenic imbalance, hypertension and improved placental and fetal weight. Taken together, our results demonstrate that procyanidin B2 inhibits sFlt-1 secretion and ameliorates endothelial dysfunction and impaired angiogenesis via the Nrf2/PPARγ axis in preeclampsia. Procyanidin B2 may be a novel therapeutic agent for treatment of preeclampsia.
Collapse
Affiliation(s)
- Lei Liu
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Rencheng Wang
- Department of Obstetrics and Gynecology, Renhe Hospital Baoshan District, Shanghai 200431, China
| | - Ran Xu
- Department of Obstetrics and Gynecology, Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, 310030, China
| | - Yuening Chu
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Weirong Gu
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China.
| |
Collapse
|
36
|
Castro KR, Prado KM, Lorenzon AR, Hoshida MS, Alves EA, Francisco RPV, Zugaib M, Marques ALX, Silva ECO, Fonseca EJS, Borbely AU, Veras MM, Bevilacqua E. Serum From Preeclamptic Women Triggers Endoplasmic Reticulum Stress Pathway and Expression of Angiogenic Factors in Trophoblast Cells. Front Physiol 2022; 12:799653. [PMID: 35185601 PMCID: PMC8855099 DOI: 10.3389/fphys.2021.799653] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/17/2021] [Indexed: 01/27/2023] Open
Abstract
Preeclampsia (PE) is a hypertensive disease of pregnancy-associated with placental cell death and endoplasmic reticulum (ER) stress. It is unknown whether systemic factors aggravate placental dysfunction. We investigated whether serum factors in pregnant women with PE activate ER stress and unfolded protein responses (UPRs) in placental explants and trophoblast cells lineage. We cultured placental explants from third-trimester term placentas from control non-preeclamptic (NPE) pregnant women with serum from women with PE or controls (NPE). In PE-treated explants, there was a significant increase in gene expression of GADD34, CHOP, and SDF2. At the protein level, GRP78, SDF2, p-eIF2α, and p-eIF2α/eIF2α ratio were also augmented in treated explants. Assays were also performed in HTR8/SV-neo trophoblast cell line to characterize the putative participation of trophoblast cells. In PE serum-treated protein levels of p-eIF2a and the ratio p-elF2 α/elF2α increased after 12 h of treatment, while the gene expression of GADD34, ATF4, and CHOP was greater than control. Increased expression of SDF2 was also detected after 24 h-cultured HTR8/SV-neo cells. PE serum increased sFLT1 gene expression and decreased PlGF gene expression in placental explants. Morphologically, PE serum increased the number of syncytial knots and reduced placental cell metabolism and viability. Analysis of the serum of pregnant women with PE through Raman spectroscopy showed changes in amino acids, carotenoids, lipids, and DNA/RNA, which may be associated with the induction of ER stress found in chorionic villi treated with this serum. In conclusion, this study provides evidence that the serum of pregnant women with PE may impact placental villi changing its morphology, viability, and secreted functional factors while triggers ER stress and an UPR. The differences between PE and control sera include molecules acting as inducing factors in these processes. In summary, the results obtained in our assays suggest that after the development of PE, the serum profile of pregnant women may be an additional factor that feeds a continuous imbalance of placental homeostasis. In addition, this study may expand the possibilities for understanding the pathogenesis of this disorder.
Collapse
Affiliation(s)
- Karla R. Castro
- Laboratory for Studies in Maternal-Fetal Interactions and Placenta, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Karen M. Prado
- Laboratory for Studies in Maternal-Fetal Interactions and Placenta, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Aline R. Lorenzon
- Laboratory for Studies in Maternal-Fetal Interactions and Placenta, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Huntington Medicina Reprodutiva—Eugin Group, São Paulo, Brazil
| | - Mara S. Hoshida
- Department of Obstetrics and Gynecology, School of Medicine, University of São Paulo – HCFMUSP, São Paulo, Brazil
| | - Eliane A. Alves
- Department of Obstetrics and Gynecology, School of Medicine, University of São Paulo – HCFMUSP, São Paulo, Brazil
| | - Rossana P. V. Francisco
- Department of Obstetrics and Gynecology, School of Medicine, University of São Paulo – HCFMUSP, São Paulo, Brazil
| | - Marcelo Zugaib
- Department of Obstetrics and Gynecology, School of Medicine, University of São Paulo – HCFMUSP, São Paulo, Brazil
| | - Aldilane L. X. Marques
- Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, Brazil
| | - Elaine C. O. Silva
- Optics and Nanoscopy Group, Institute of Physics, Federal University of Alagoas, Maceio, Brazil
| | - Eduardo J. S. Fonseca
- Optics and Nanoscopy Group, Institute of Physics, Federal University of Alagoas, Maceio, Brazil
| | - Alexandre U. Borbely
- Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, Brazil
| | - Mariana M. Veras
- Laboratory of Experimental Air Pollution, Department of Pathology, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Estela Bevilacqua
- Laboratory for Studies in Maternal-Fetal Interactions and Placenta, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
37
|
Huovinen M, Ietta F, Repo J, Paulesu L, Vähäkangas K. The effect of ethanol and nicotine on ER stress in human placental villous explants. Curr Res Toxicol 2022; 3:100081. [PMID: 35814289 PMCID: PMC9256831 DOI: 10.1016/j.crtox.2022.100081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 06/03/2022] [Accepted: 06/24/2022] [Indexed: 11/29/2022] Open
Abstract
Nicotine increased the GRP78/BiP protein in first trimester and term placental villous explants. Nicotine can cause endoplasmic reticulum stress in human placenta. Placental villous explants can be isolated from first trimester and term placenta to compare responses to toxic compounds.
Pregnant mothers continue smoking and drinking during pregnancy. To clarify the mechanisms of nicotine and ethanol toxicity during development, we have examined their effects on endoplasmic reticulum (ER) stress in human first trimester and term placental explants. First trimester and term human placental explants were treated with ethanol (2 ‰) or nicotine (15 µM), or their combination. The ER stress markers glucose regulated protein 78 (GRP78/BiP) and inositol requiring enzyme 1 α (IRE1α) were analyzed by immunoblotting. A statistically significant increase (p < 0.05) of GRP78/BiP by nicotine was noted in first trimester placental explants at 48 h, and in term placental explants at 24 h. Ethanol did not change protein expression of GRP78/BiP in either first trimester or term placental explants. IRE1α increased, although not statistically significantly, by all treatments in both first trimester and term placental explants. Thus, regardless of the known structural and functional differences in early and late placenta, both responded very similarly to the toxic compounds studied. These data support our earlier results in BeWo cells (Repo et al., 2014) implicating that nicotine induces ER stress in human placenta and may interfere with placental functions potentially disrupting fetal growth and development.
Collapse
|
38
|
Deval G, Boland S, Fournier T, Ferecatu I. On Placental Toxicology Studies and Cerium Dioxide Nanoparticles. Int J Mol Sci 2021; 22:ijms222212266. [PMID: 34830142 PMCID: PMC8624015 DOI: 10.3390/ijms222212266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/20/2021] [Accepted: 11/08/2021] [Indexed: 12/31/2022] Open
Abstract
The human placenta is a transient organ essential for pregnancy maintenance, fetal development and growth. It has several functions, including that of a selective barrier against pathogens and xenobiotics from maternal blood. However, some pollutants can accumulate in the placenta or pass through with possible repercussions on pregnancy outcomes. Cerium dioxide nanoparticles (CeO2 NPs), also termed nanoceria, are an emerging pollutant whose impact on pregnancy is starting to be defined. CeO2 NPs are already used in different fields for industrial and commercial applications and have even been proposed for some biomedical applications. Since 2010, nanoceria have been subject to priority monitoring by the Organization for Economic Co-operation and Development in order to assess their toxicity. This review aims to summarize the current methods and models used for toxicology studies on the placental barrier, from the basic ones to the very latest, as well as to overview the most recent knowledge of the impact of CeO2 NPs on human health, and more specifically during the sensitive window of pregnancy. Further research is needed to highlight the relationship between environmental exposure to CeO2 and placental dysfunction with its implications for pregnancy outcome.
Collapse
Affiliation(s)
- Gaëlle Deval
- Université de Paris, Inserm, UMR-S 1139, 3PHM, Faculté de Pharmacie, 75006 Paris, France; (G.D.); (T.F.)
| | - Sonja Boland
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France;
| | - Thierry Fournier
- Université de Paris, Inserm, UMR-S 1139, 3PHM, Faculté de Pharmacie, 75006 Paris, France; (G.D.); (T.F.)
| | - Ioana Ferecatu
- Université de Paris, Inserm, UMR-S 1139, 3PHM, Faculté de Pharmacie, 75006 Paris, France; (G.D.); (T.F.)
- Correspondence: ; Tel.: +33-1-5373-9605
| |
Collapse
|
39
|
Manzan-Martins C, Paulesu L. Impact of bisphenol A (BPA) on cells and tissues at the human materno-fetal interface. Tissue Cell 2021; 73:101662. [PMID: 34628212 DOI: 10.1016/j.tice.2021.101662] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 02/07/2023]
Abstract
Bisphenol A (BPA) is an endocrine disruptor extensively used in the production of polycarbonate plastics and epoxy resins and a component of liquid and food containers. It is a hazard in the prenatal period because of its presence in the placenta, fetal membranes, amniotic fluid, maternal and fetal blood and its ability to cross the placenta and reach the fetus. Estimation of the risk of BPA exposure during in utero life is extremely important in order to prevent complications of pregnancy and fetal growth. This review describes in vitro models of the human materno-fetal interface. It also outlines the effects of BPA at doses indicated as "physiological", namely at the concentrations found in the general population, and at "supraphysiological" and "subphysiological" doses, i.e. above and below the physiological range. This work will help clarify the discrepancies observed in studies on the effects of BPA on human reproduction and pregnancy, and it will be useful for the choice of appropriate in vitro models for future studies aimed at identifying the potential impact of BPA on specific functional processes.
Collapse
Affiliation(s)
| | - L Paulesu
- Department of Life Sciences, University of Siena, Siena, Italy.
| |
Collapse
|
40
|
Schmidt A, Schmidt A, Markert UR. The road (not) taken - Placental transfer and interspecies differences. Placenta 2021; 115:70-77. [PMID: 34562829 DOI: 10.1016/j.placenta.2021.09.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 07/08/2021] [Accepted: 09/13/2021] [Indexed: 12/31/2022]
Abstract
Species differences are among the main reasons for the high failure rate of preclinical studies. A better awareness and understanding of these differences might help to improve the outcome of preclinical research. In reproduction, the placenta is the central organ regulating fetal exposure to a substance circulating in the maternal organism. Exact information about placental transfer can help to better estimate the toxic potential of a substance. From an evolutionary point of view, the chorioallantoic placenta is the organ with the highest anatomical diversity among species. Moreover, frequently used animal models in reproduction belong to rodents and lagomorphs, two groups that are characterized by the generation of an additional type of placenta, which is crucial for fetal development, but absent from humans: the inverted yolk sac placenta. Taken together, the translatability of placental transfer studies from laboratory animals to humans is challenging, which is supported by the fact that numerous species-dependent toxic effects are described in literature. Thus, reliable human-relevant data are frequently lacking and the toxic potential of chemicals and pharmaceuticals for humans can hardly be estimated, often resulting in recommendations that medical treatments or exposure to chemicals should be avoided for safety reasons. Although species differences of placental anatomy have been described frequently and the need for human-relevant research models has been emphasized, analyses of substances with species-dependent placental transfer have been performed only sporadically. Here, we present examples for species-specific placental transfer, including that of nanoparticles and pharmaceuticals, and discuss potential underlying mechanisms. With respect to the COVID 19-pandemic it might be of interest that some antiviral drugs are reported to feature species-specific placental transfer. Further, differences in placental structure and antibody transfer may affect placental transfer of ZIKA virus.
Collapse
Affiliation(s)
- André Schmidt
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Am Klinikum 1, 07747, Jena, Germany.
| | - Astrid Schmidt
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Udo R Markert
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Am Klinikum 1, 07747, Jena, Germany
| |
Collapse
|
41
|
The impact of Zika virus exposure on the placental proteomic profile. Biochim Biophys Acta Mol Basis Dis 2021; 1868:166270. [PMID: 34582966 DOI: 10.1016/j.bbadis.2021.166270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/12/2021] [Accepted: 09/12/2021] [Indexed: 11/21/2022]
Abstract
Zika virus (ZIKV) infection has caused severe unexpected clinical outcomes in neonates and adults during the recent outbreak in Latin America, particularly in Brazil. Congenital malformations associated with ZIKV have been frequently reported; nevertheless, the mechanism of vertical transmission and the involvement of placental cells remains unclear. In this study, we applied quantitative proteomics analysis in a floating explant model of chorionic villi of human placental tissues incubated with ZIKV and with ZIKV pre-adsorbed with anti-ZIKV envelope protein. Proteomic data are available via ProteomeXchange with identifier PXD025764. Altered levels of proteins were involved in cell proliferation, apoptosis, inflammatory processes, and the integrin-cytoskeleton complex. Antibody-opsonized ZIKV particles differentially modulated the pattern of protein expression in placental cells; this phenomenon may play a pivotal role in determining the course of infection and the role of mixed infections. The expression of specific proteins was also evaluated by immunoperoxidase assays. These data fill gaps in our understanding of early events after ZIKV placental exposure and help identify infection control targets.
Collapse
|
42
|
de Alwis N, Beard S, Binder NK, Pritchard N, Kaitu'u-Lino TJ, Walker SP, Stock O, Groom KM, Petersen S, Henry A, Said JM, Seeho S, Kane SC, Tong S, Hannan NJ. LOX-1 expression is reduced in placenta from pregnancies complicated by preeclampsia and in hypoxic cytotrophoblast. Pregnancy Hypertens 2021; 25:255-261. [PMID: 34325289 DOI: 10.1016/j.preghy.2021.07.243] [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: 04/23/2021] [Revised: 07/03/2021] [Accepted: 07/19/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVES The lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is upregulated in the maternal vasculature in preeclampsia, and contributes to oxidative stress and endothelial dysfunction. However, its function in the placenta is unclear. This paper investigated LOX-1 expression in models of placental dysfunction and preeclampsia, and whether candidate therapeutics for preeclampsia could alter its expression. STUDY DESIGN Placentas were collected from preterm pregnancies and cases of preterm preeclampsia and fetal growth restriction. Blood was collected from participants whose pregnancies were complicated by preterm fetal growth restriction and/or preeclampsia. Primary cytotrophoblast and placental explant tissue were cultured under hypoxic (1% O2) or normoxic (8% O2) conditions. Cytotrophoblast were exposed to 10% preeclamptic or control serum. Cytotrophoblast and preeclamptic explant tissue were treated with 100 µM esomeprazole, lansoprazole or rabeprazole. MAIN OUTCOME MEASURES LOX-1 expression was assessed in all samples via qPCR. RESULTS LOX-1 expression was reduced in placentas from cases of preterm preeclampsia, but not fetal growth restriction, compared to controls. LOX-1 expression was reduced in cytotrophoblast under hypoxia, but not in explant tissue. Treatment with preeclamptic serum in vitro did not alter cytotrophoblast LOX-1 expression. Circulating LOX-1 mRNA was unaltered in patients with fetal growth restriction, preeclampsia, and fetal hypoxia, compared to controls. Treatment with esomeprazole or lansoprazole in vitro increased placental LOX-1 expression. CONCLUSIONS LOX-1 expression is reduced in preeclamptic placentas and hypoxic cytotrophoblast. Esomeprazole and lansoprazole increase placental LOX-1 expression. These findings demonstrate a role for LOX-1 in the placenta, and improve our understanding of maternal adaptations in pregnancy complications.
Collapse
Affiliation(s)
- Natasha de Alwis
- Therapeutics Discovery and Vascular Function in Pregnancy Group, Mercy Hospital for Women, Heidelberg, Victoria 3084, Australia; Translational Obstetrics Group, Mercy Hospital for Women, Heidelberg, Victoria 3084, Australia; Mercy Perinatal, Mercy Hospital for Women, Heidelberg 3084, Victoria, Australia; Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - Sally Beard
- Therapeutics Discovery and Vascular Function in Pregnancy Group, Mercy Hospital for Women, Heidelberg, Victoria 3084, Australia; Translational Obstetrics Group, Mercy Hospital for Women, Heidelberg, Victoria 3084, Australia; Mercy Perinatal, Mercy Hospital for Women, Heidelberg 3084, Victoria, Australia; Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - Natalie K Binder
- Therapeutics Discovery and Vascular Function in Pregnancy Group, Mercy Hospital for Women, Heidelberg, Victoria 3084, Australia; Translational Obstetrics Group, Mercy Hospital for Women, Heidelberg, Victoria 3084, Australia; Mercy Perinatal, Mercy Hospital for Women, Heidelberg 3084, Victoria, Australia; Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - Natasha Pritchard
- Translational Obstetrics Group, Mercy Hospital for Women, Heidelberg, Victoria 3084, Australia; Mercy Perinatal, Mercy Hospital for Women, Heidelberg 3084, Victoria, Australia; Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Translational Obstetrics Group, Mercy Hospital for Women, Heidelberg, Victoria 3084, Australia; Mercy Perinatal, Mercy Hospital for Women, Heidelberg 3084, Victoria, Australia; Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - Susan P Walker
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg 3084, Victoria, Australia; Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - Owen Stock
- Translational Obstetrics Group, Mercy Hospital for Women, Heidelberg, Victoria 3084, Australia; Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - Katie M Groom
- Liggins Institute, University of Auckland, Auckland 1023, New Zealand
| | - Scott Petersen
- Centre for Maternal Fetal Medicine, Mater Mothers' Hospital, South Brisbane 4101, Queensland, Australia
| | - Amanda Henry
- School of Women's and Children's Health, UNSW Medicine, University of New South Wales, Sydney, Australia
| | - Joanne M Said
- Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia; Maternal Fetal Medicine, Joan Kirner Women's & Children's Sunshine Hospital, St Albans 3021, Victoria, Australia
| | - Sean Seeho
- The University of Sydney Northern Clinical School, Women and Babies Research, St Leonards 2065, New South Wales, Australia
| | - Stefan C Kane
- Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia; Department of Maternal Fetal Medicine, Royal Women's Hospital, Parkville 3052, Victoria, Australia
| | - Stephen Tong
- Translational Obstetrics Group, Mercy Hospital for Women, Heidelberg, Victoria 3084, Australia; Mercy Perinatal, Mercy Hospital for Women, Heidelberg 3084, Victoria, Australia; Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - Natalie J Hannan
- Therapeutics Discovery and Vascular Function in Pregnancy Group, Mercy Hospital for Women, Heidelberg, Victoria 3084, Australia; Translational Obstetrics Group, Mercy Hospital for Women, Heidelberg, Victoria 3084, Australia; Mercy Perinatal, Mercy Hospital for Women, Heidelberg 3084, Victoria, Australia; Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia.
| |
Collapse
|
43
|
Ayala-Ramírez P, Machuca-Acevedo C, Gámez T, Quijano S, Barreto A, Silva JL, Olaya-C M, García-Robles R. Assessment of Placental Extracellular Vesicles-Associated Fas Ligand and TNF-Related Apoptosis-Inducing Ligand in Pregnancies Complicated by Early and Late Onset Preeclampsia. Front Physiol 2021; 12:708824. [PMID: 34366896 PMCID: PMC8342945 DOI: 10.3389/fphys.2021.708824] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
Preeclampsia (PE) is a hypertensive disorder that affects 2-8% of pregnancies and is one of the main causes of fetal, neonatal, and maternal mortality and morbidity worldwide. Although PE etiology and pathophysiology remain unknown, there is evidence that the hyperactivation of maternal immunity cells against placental cells triggers trophoblast cell apoptosis and death. It has also been reported that placenta-derived extracellular vesicles (EV) carry Fas ligand (FasL) and Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and trigger apoptosis in Jurkat T cells. This study aimed to quantify and compare FasL and TRAIL expression in EV derived from cultures of placenta explants from women with PE (early versus late) and women with uncomplicated pregnancies. Also, the study assessed EV capacity to induce apoptosis in Jurkat T cells. The authors isolated EV from placenta explant cultures, quantified FasL and TRAIL using ELISA, and analyzed EV apoptosis-inducing capability by flow cytometry. Results showed increased FasL and TRAIL in EV derived from placenta of women with PE, and increased EV apoptosis-inducing capability in Jurkat T cells. These results offer supporting evidence that EV FasL and TRAIL play a role in the pathophysiology of PE.
Collapse
Affiliation(s)
- Paola Ayala-Ramírez
- Human Genetics Institute, Faculty of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Catalina Machuca-Acevedo
- Human Genetics Institute, Faculty of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Tatiana Gámez
- Human Genetics Institute, Faculty of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Sandra Quijano
- Grupo de Inmunobiología y Biología Celular, Unidad de Investigación en Ciencias Biomédicas, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Alfonso Barreto
- Grupo de Inmunobiología y Biología Celular, Unidad de Investigación en Ciencias Biomédicas, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Jaime L. Silva
- Department of Obstetrics and Gynecology, Faculty of Medicine, Hospital Universitario San Ignacio, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Mercedes Olaya-C
- Department of Pathology, Faculty of Medicine, Hospital Universitario San Ignacio, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Reggie García-Robles
- Department of Physiological Sciences, Faculty of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
44
|
Wong MK, Li EW, Adam M, Selvaganapathy PR, Raha S. Establishment of an in vitro placental barrier model cultured under physiologically relevant oxygen levels. Mol Hum Reprod 2021; 26:353-365. [PMID: 32159799 DOI: 10.1093/molehr/gaaa018] [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: 12/11/2019] [Revised: 02/05/2020] [Indexed: 12/16/2022] Open
Abstract
The human placental barrier facilitates many key functions during pregnancy, most notably the exchange of all substances between the mother and fetus. However, preclinical models of the placental barrier often lacked the multiple cell layers, syncytialization of the trophoblast cells and the low oxygen levels that are present within the body. Therefore, we aimed to design and develop an in vitro model of the placental barrier that would reinstate these factors and enable improved investigations of barrier function. BeWo placental trophoblastic cells and human umbilical vein endothelial cells were co-cultured on contralateral sides of an extracellular matrix-coated transwell insert to establish a multilayered barrier. Epidermal growth factor and forskolin led to significantly increased multi-nucleation of the BeWo cell layer and increased biochemical markers of syncytial fusion, for example syncytin-1 and hCGβ. Our in vitro placental barrier possessed size-specific permeability, with 4000-Da molecules experiencing greater transport and a lower apparent permeability coefficient than 70 000-Da molecules. We further demonstrated that the BeWo layer had greater resistance to smaller molecules compared to the endothelial layer. Chronic, physiologically low oxygen exposure (3-8%) increased the expression of hypoxia-inducible factor 1α and syncytin-1, further increased multi-nucleation of the BeWo cell layer and decreased barrier permeability only against smaller molecules (457 Da/4000 Da). In conclusion, we built a novel in vitro co-culture model of the placental barrier that possessed size-specific permeability and could function under physiologically low oxygen levels. Importantly, this will enable future researchers to better study the maternal-fetal transport of nutrients and drugs during pregnancy.
Collapse
Affiliation(s)
- Michael K Wong
- Graduate Program of Medical Science, McMaster University, Hamilton, Ontario, Canada.,Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Edward W Li
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Mohamed Adam
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | - Sandeep Raha
- Graduate Program of Medical Science, McMaster University, Hamilton, Ontario, Canada.,Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada.,Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| |
Collapse
|
45
|
Kupper N, Pritz E, Siwetz M, Guettler J, Huppertz B. Placental Villous Explant Culture 2.0: Flow Culture Allows Studies Closer to the In Vivo Situation. Int J Mol Sci 2021; 22:ijms22147464. [PMID: 34299084 PMCID: PMC8308011 DOI: 10.3390/ijms22147464] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/09/2021] [Indexed: 01/15/2023] Open
Abstract
During pregnancy, freely floating placental villi are adapted to fluid shear stress due to placental perfusion with maternal plasma and blood. In vitro culture of placental villous explants is widely performed under static conditions, hoping the conditions may represent the in utero environment. However, static placental villous explant culture dramatically differs from the in vivo situation. Thus, we established a flow culture system for placental villous explants and compared commonly used static cultured tissue to flow cultured tissue using transmission and scanning electron microscopy, immunohistochemistry, and lactate dehydrogenase (LDH) and human chorionic gonadotropin (hCG) measurements. The data revealed a better structural and biochemical integrity of flow cultured tissue compared to static cultured tissue. Thus, this new flow system can be used to simulate the blood flow from the mother to the placenta and back in the most native-like in vitro system so far and thus can enable novel study designs.
Collapse
|
46
|
Jaremek A, Jeyarajah MJ, Jaju Bhattad G, Renaud SJ. Omics Approaches to Study Formation and Function of Human Placental Syncytiotrophoblast. Front Cell Dev Biol 2021; 9:674162. [PMID: 34211975 PMCID: PMC8240757 DOI: 10.3389/fcell.2021.674162] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/24/2021] [Indexed: 01/12/2023] Open
Abstract
Proper development of the placenta is vital for pregnancy success. The placenta regulates exchange of nutrients and gases between maternal and fetal blood and produces hormones essential to maintain pregnancy. The placental cell lineage primarily responsible for performing these functions is a multinucleated entity called syncytiotrophoblast. Syncytiotrophoblast is continuously replenished throughout pregnancy by fusion of underlying progenitor cells called cytotrophoblasts. Dysregulated syncytiotrophoblast formation disrupts the integrity of the placental exchange surface, which can be detrimental to maternal and fetal health. Moreover, various factors produced by syncytiotrophoblast enter into maternal circulation, where they profoundly impact maternal physiology and are promising diagnostic indicators of pregnancy health. Despite the multifunctional importance of syncytiotrophoblast for pregnancy success, there is still much to learn about how its formation is regulated in normal and diseased states. ‘Omics’ approaches are gaining traction in many fields to provide a more holistic perspective of cell, tissue, and organ function. Herein, we review human syncytiotrophoblast development and current model systems used for its study, discuss how ‘omics’ strategies have been used to provide multidimensional insights into its formation and function, and highlight limitations of current platforms as well as consider future avenues for exploration.
Collapse
Affiliation(s)
- Adam Jaremek
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Mariyan J Jeyarajah
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Gargi Jaju Bhattad
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Stephen J Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Children's Health Research Institute, Lawson Health Research Institute, London, ON, Canada
| |
Collapse
|
47
|
Liu Chung Ming C, Sesperez K, Ben-Sefer E, Arpon D, McGrath K, McClements L, Gentile C. Considerations to Model Heart Disease in Women with Preeclampsia and Cardiovascular Disease. Cells 2021; 10:899. [PMID: 33919808 PMCID: PMC8070848 DOI: 10.3390/cells10040899] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/11/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
Preeclampsia is a multifactorial cardiovascular disorder diagnosed after 20 weeks of gestation, and is the leading cause of death for both mothers and babies in pregnancy. The pathophysiology remains poorly understood due to the variability and unpredictability of disease manifestation when studied in animal models. After preeclampsia, both mothers and offspring have a higher risk of cardiovascular disease (CVD), including myocardial infarction or heart attack and heart failure (HF). Myocardial infarction is an acute myocardial damage that can be treated through reperfusion; however, this therapeutic approach leads to ischemic/reperfusion injury (IRI), often leading to HF. In this review, we compared the current in vivo, in vitro and ex vivo model systems used to study preeclampsia, IRI and HF. Future studies aiming at evaluating CVD in preeclampsia patients could benefit from novel models that better mimic the complex scenario described in this article.
Collapse
Affiliation(s)
- Clara Liu Chung Ming
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Sydney, NSW 2007, Australia; (C.L.C.M.); (E.B.-S.); (D.A.)
| | - Kimberly Sesperez
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (K.S.); (K.M.); (L.M.)
| | - Eitan Ben-Sefer
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Sydney, NSW 2007, Australia; (C.L.C.M.); (E.B.-S.); (D.A.)
| | - David Arpon
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Sydney, NSW 2007, Australia; (C.L.C.M.); (E.B.-S.); (D.A.)
| | - Kristine McGrath
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (K.S.); (K.M.); (L.M.)
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (K.S.); (K.M.); (L.M.)
| | - Carmine Gentile
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Sydney, NSW 2007, Australia; (C.L.C.M.); (E.B.-S.); (D.A.)
- Sydney Medical School, The University of Sydney, Sydney, NSW 2000, Australia
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
48
|
Syncytiotrophoblast Derived Extracellular Vesicles in Relation to Preeclampsia. MATERNAL-FETAL MEDICINE 2021. [DOI: 10.1097/fm9.0000000000000093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
49
|
Hypoxia and oxidative stress induce sterile placental inflammation in vitro. Sci Rep 2021; 11:7281. [PMID: 33790316 PMCID: PMC8012380 DOI: 10.1038/s41598-021-86268-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/08/2021] [Indexed: 12/18/2022] Open
Abstract
Fetal growth restriction (FGR) and stillbirth are associated with placental dysfunction and inflammation and hypoxia, oxidative and nitrative stress are implicated in placental damage. Damage-associated molecular patterns (DAMPs) are elevated in pregnancies at increased risk of FGR and stillbirth and are associated with increase in pro-inflammatory placental cytokines. We hypothesised that placental insults lead to release of DAMPs, promoting placental inflammation. Placental tissue from uncomplicated pregnancies was exposed in vitro to hypoxia, oxidative or nitrative stress. Tissue production and release of DAMPs and cytokines was determined. Oxidative stress and hypoxia caused differential release of DAMPs including uric acid, HMGB1, S100A8, cell-free fetal DNA, S100A12 and HSP70. After oxidative stress pro-inflammatory cytokines (IL-1α, IL-1β, IL-6, IL-8, TNFα, CCL2) were increased both within explants and in conditioned culture medium. Hypoxia increased tissue IL-1α/β, IL-6, IL-8 and TNFα levels, and release of IL-1α, IL-6 and IL-8, whereas CCL2 and IL-10 were reduced. IL1 receptor antagonist (IL1Ra) treatment prevented hypoxia- and oxidative stress-induced IL-6 and IL-8 release. These findings provide evidence that relevant stressors induce a sterile inflammatory profile in placental tissue which can be partially blocked by IL1Ra suggesting this agent has translational potential to prevent placental inflammation evident in FGR and stillbirth.
Collapse
|
50
|
Massimiani M, Tiralongo GM, Salvi S, Fruci S, Lacconi V, La Civita F, Mancini M, Stuhlmann H, Valensise H, Campagnolo L. Treatment of pregnancies complicated by intrauterine growth restriction with nitric oxide donors increases placental expression of Epidermal Growth Factor-Like Domain 7 and improves fetal growth: A pilot study. Transl Res 2021; 228:28-41. [PMID: 32784003 DOI: 10.1016/j.trsl.2020.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 11/25/2022]
Abstract
Intrauterine growth restriction (IUGR) is a pathological condition of pregnancy with high perinatal mortality and morbidity, characterized by inadequate fetal growth associated to altered maternal hemodynamics with impaired uteroplacental blood flow and placental insufficiency. To date, iatrogenic premature delivery remains the elective therapeutic strategy. However, in recent years the possibility of a therapeutic approach with vasodilators and myorelaxants, such as nitric oxide (NO) donors, has gained interest. NO controls many endothelial cell functions, including angiogenesis and vascular permeability, by regulating the expression of angiogenic factors, such as Vascular Endothelial Growth Factor. In the present study, we investigated if treatment of pregnancies complicated by IUGR with NO donors affects the expression of Epidermal Growth Factor-Like Domain 7 (EGFL7), a secreted endothelial factor, previously demonstrated to be expressed by both endothelial and trophoblast cells and involved in proper placental development. NO donor treatment induced placental levels of EGFL7 and, in association with oral fluids, significantly improved fetal growth. Ex vivo experiments confirmed that NO donors increased expression and secretion of EGFL7 by villous explants. To specifically investigate the potential response of trophoblast cells to NO, we treated HTR8-sVneo cells with NO donors and observed induction of EGFL7 expression. Altogether, our findings indicate that NO induces endothelial and trophoblast expression of EGFL7 in the placenta and improves fetal growth, suggesting a correlation between placental levels of EGFL7 and pregnancy outcome.
Collapse
Affiliation(s)
- Micol Massimiani
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy; Saint Camillus International University of Health Sciences, Rome, Italy
| | - Grazia M Tiralongo
- Department of Obstetrics and Gynecology, Casilino General Hospital, Rome, Italy
| | - Silvia Salvi
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, UOC di Patologia Ostetrica, Fondazione Policlinico Universitario "A Gemelli" IRCCS, Rome, Italy
| | | | - Valentina Lacconi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Fabio La Civita
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Marta Mancini
- Department of Obstetrics and Gynecology, Casilino General Hospital, Rome, Italy
| | - Heidi Stuhlmann
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York
| | - Herbert Valensise
- Department of Obstetrics and Gynecology, Casilino General Hospital, Rome, Italy; Obstetrics and Gynecology Unit, Department of Surgical Sciences, University of Rome Tor Vergata, Rome, Italy.
| | - Luisa Campagnolo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|