51
|
Morey R, Farah O, Kallol S, Requena DF, Meads M, Moretto-Zita M, Soncin F, Laurent LC, Parast MM. Transcriptomic Drivers of Differentiation, Maturation, and Polyploidy in Human Extravillous Trophoblast. Front Cell Dev Biol 2021; 9:702046. [PMID: 34540826 PMCID: PMC8446284 DOI: 10.3389/fcell.2021.702046] [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: 04/29/2021] [Accepted: 07/27/2021] [Indexed: 12/20/2022] Open
Abstract
During pregnancy, conceptus-derived extravillous trophoblast (EVT) invades the endomyometrium, anchors the placenta to the maternal uterus, and remodels the spiral arteries in order to establish maternal blood supply to the fetoplacental unit. Recent reports have described early gestation EVT as polyploid and senescent. Here, we extend these reports by performing comprehensive profiling of both the genomic organization and transcriptome of first trimester and term EVT. We define pathways and gene regulatory networks involved in both initial differentiation and maturation of this important trophoblast lineage at the maternal-fetal interface. Our results suggest that like first trimester EVT, term EVT undergoes senescence and endoreduplication, is primarily tetraploid, and lacks high rates of copy number variations. Additionally, we have highlighted senescence and polyploidy-related genes, pathways, networks, and transcription factors that appeared to be important in normal EVT differentiation and maturation and validated a key role for the unfolded protein response in this context.
Collapse
Affiliation(s)
- Robert Morey
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States.,Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal-Fetal Medicine, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Omar Farah
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Sampada Kallol
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Daniela F Requena
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Morgan Meads
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Matteo Moretto-Zita
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Francesca Soncin
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Louise C Laurent
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal-Fetal Medicine, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Mana M Parast
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States.,Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
52
|
The Impact of Hypoxia in Early Pregnancy on Placental Cells. Int J Mol Sci 2021; 22:ijms22189675. [PMID: 34575844 PMCID: PMC8466283 DOI: 10.3390/ijms22189675] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/04/2021] [Accepted: 09/05/2021] [Indexed: 12/14/2022] Open
Abstract
Oxygen levels in the placental microenvironment throughout gestation are not constant, with severe hypoxic conditions present during the first trimester. This hypoxic phase overlaps with the most critical stages of placental development, i.e., blastocyst implantation, cytotrophoblast invasion, and spiral artery remodeling initiation. Dysregulation of any of these steps in early gestation can result in pregnancy loss and/or adverse pregnancy outcomes. Hypoxia has been shown to regulate not only the self-renewal, proliferation, and differentiation of trophoblast stem cells and progenitor cells, but also the recruitment, phenotype, and function of maternal immune cells. In this review, we will summarize how oxygen levels in early placental development determine the survival, fate, and function of several important cell types, e.g., trophoblast stem cells, extravillous trophoblasts, syncytiotrophoblasts, uterine natural killer cells, Hofbauer cells, and decidual macrophages. We will also discuss the cellular mechanisms used to cope with low oxygen tensions, such as the induction of hypoxia-inducible factor (HIF) or mammalian target of rapamycin (mTOR) signals, regulation of the metabolic pathway, and adaptation to autophagy. Understanding the beneficial roles of hypoxia in early placental development will provide insights into the root cause(s) of some pregnancy disorders, such as spontaneous abortion, preeclampsia, and intrauterine growth restriction.
Collapse
|
53
|
ASCL2 reciprocally controls key trophoblast lineage decisions during hemochorial placenta development. Proc Natl Acad Sci U S A 2021; 118:2016517118. [PMID: 33649217 DOI: 10.1073/pnas.2016517118] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Invasive trophoblast cells are critical to spiral artery remodeling in hemochorial placentation. Insufficient trophoblast cell invasion and vascular remodeling can lead to pregnancy disorders including preeclampsia, preterm birth, and intrauterine growth restriction. Previous studies in mice identified achaete-scute homolog 2 (ASCL2) as essential to extraembryonic development. We hypothesized that ASCL2 is a critical and conserved regulator of invasive trophoblast cell lineage development. In contrast to the mouse, the rat possesses deep intrauterine trophoblast cell invasion and spiral artery remodeling similar to human placentation. In this study, we investigated invasive/extravillous trophoblast (EVT) cell differentiation using human trophoblast stem (TS) cells and a loss-of-function mutant Ascl2 rat model. ASCL2 transcripts are expressed in the EVT column and junctional zone, which represent tissue sources of invasive trophoblast progenitor cells within human and rat placentation sites, respectively. Differentiation of human TS cells into EVT cells resulted in significant up-regulation of ASCL2 and several other transcripts indicative of EVT cell differentiation. Disruption of ASCL2 impaired EVT cell differentiation, as indicated by cell morphology and transcript profiles. RNA sequencing analysis of ASCL2-deficient trophoblast cells identified both down-regulation of EVT cell-associated transcripts and up-regulation of syncytiotrophoblast-associated transcripts, indicative of dual activating and repressing functions. ASCL2 deficiency in the rat impacted placental morphogenesis, resulting in junctional zone dysgenesis and failed intrauterine trophoblast cell invasion. ASCL2 acts as a critical and conserved regulator of invasive trophoblast cell lineage development and a modulator of the syncytiotrophoblast lineage.
Collapse
|
54
|
Proteome profiling of human placenta reveals developmental stage-dependent alterations in protein signature. Clin Proteomics 2021; 18:18. [PMID: 34372761 PMCID: PMC8351416 DOI: 10.1186/s12014-021-09324-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 08/01/2021] [Indexed: 12/26/2022] Open
Abstract
Introduction Placenta is a complex organ that plays a significant role in the maintenance of pregnancy health. It is a dynamic organ that undergoes dramatic changes in growth and development at different stages of gestation. In the first-trimester, the conceptus develops in a low oxygen environment that favors organogenesis in the embryo and cell proliferation and angiogenesis in the placenta; later in pregnancy, higher oxygen concentration is required to support the rapid growth of the fetus. This oxygen transition, which appears unique to the human placenta, must be finely tuned through successive rounds of protein signature alterations. This study compares placental proteome in normal first-trimester (FT) and term human placentas (TP). Methods Normal human first-trimester and term placental samples were collected and differentially expressed proteins were identified using two-dimensional liquid chromatography-tandem mass spectrometry. Results Despite the overall similarities, 120 proteins were differently expressed in first and term placentas. Out of these, 72 were up-regulated and 48 were down-regulated in the first when compared with the full term placentas. Twenty out of 120 differently expressed proteins were sequenced, among them seven showed increased (GRP78, PDIA3, ENOA, ECH1, PRDX4, ERP29, ECHM), eleven decreased (TRFE, ALBU, K2C1, ACTG, CSH2, PRDX2, FABP5, HBG1, FABP4, K2C8, K1C9) expression in first-trimester compared to the full-term placentas and two proteins exclusively expressed in first-trimester placentas (MESD, MYDGF). Conclusion According to Reactome and PANTHER softwares, these proteins were mostly involved in response to chemical stimulus and stress, regulation of biological quality, programmed cell death, hemostatic and catabolic processes, protein folding, cellular oxidant detoxification, coagulation and retina homeostasis. Elucidation of alteration in protein signature during placental development would provide researchers with a better understanding of the critical biological processes of placentogenesis and delineate proteins involved in regulation of placental function during development. Supplementary Information The online version contains supplementary material available at 10.1186/s12014-021-09324-y.
Collapse
|
55
|
Hypoxia and the integrated stress response promote pulmonary hypertension and preeclampsia: Implications in drug development. Drug Discov Today 2021; 26:2754-2773. [PMID: 34302972 DOI: 10.1016/j.drudis.2021.07.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/31/2021] [Accepted: 07/14/2021] [Indexed: 11/21/2022]
Abstract
Chronic hypoxia is a common cause of pulmonary hypertension, preeclampsia, and intrauterine growth restriction (IUGR). The molecular mechanisms underlying these diseases are not completely understood. Chronic hypoxia may induce the generation of reactive oxygen species (ROS) in mitochondria, promote endoplasmic reticulum (ER) stress, and result in the integrated stress response (ISR) in the pulmonary artery and uteroplacental tissues. Numerous studies have implicated hypoxia-inducible factors (HIFs), oxidative stress, and ER stress/unfolded protein response (UPR) in the development of pulmonary hypertension, preeclampsia and IUGR. This review highlights the roles of HIFs, mitochondria-derived ROS and UPR, as well as their interplay, in the pathogenesis of pulmonary hypertension and preeclampsia, and their implications in drug development.
Collapse
|
56
|
Ischemia-Modified Albumin: Origins and Clinical Implications. DISEASE MARKERS 2021; 2021:9945424. [PMID: 34336009 PMCID: PMC8315882 DOI: 10.1155/2021/9945424] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/02/2021] [Accepted: 06/29/2021] [Indexed: 11/18/2022]
Abstract
Albumin is one of the most abundant proteins in the body of mammals: about 40% of its pool is located in the intravascular space and the remainder is found in the interstitial space. The content of this multifunctional protein in blood is about 60-65% of total plasma proteins. A decrease in its synthesis or changes of functional activity can destabilize oncotic blood pressure, cause a violation of transporting hormones, fatty acids, metals, and drugs. Albumin properties change under ischemic attacks associated with oxidative stress, production of reactive oxygen species, and acidosis. Under these conditions, ischemia-modified albumin (IMA) is generated that has a reduced metal-binding capacity, especially for transition metals, such as copper, nickel, and cobalt. The method of determining the cobalt-binding capability of HSA was initially proposed to evaluate IMA level and then licensed as an ACB test for routine clinical analysis for myocardial ischemia. Subsequent studies have shown the viability of the ACB test in diagnosing other diseases associated with the development of oxidative stress. This review examines recent data on IMA generation mechanisms, describes principles, advantages, and limitations of methods for evaluation of IMA levels, and provides detailed analysis of its use in diagnostic and monitoring therapeutic efficacy in different diseases.
Collapse
|
57
|
Screening Candidate Genes Regulating Placental Development from Trophoblast Transcriptome at Early Pregnancy in Dazu Black Goats ( Capra hircus). Animals (Basel) 2021; 11:ani11072132. [PMID: 34359260 PMCID: PMC8300351 DOI: 10.3390/ani11072132] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary The trophoblast is an original placental tissue whose normal proliferation, differentiation, migration, adhesion, and angiopoiesis are essential for placenta formation and fetal survival during early pregnancy. However, the key genes and molecular mechanisms involved in placenta development in goats are unknown. Herein, the morphology and histological structures of trophoblast tissues from day 20 to 30 of pregnancy were determined. RNA-sequencing was used to screen potential functional genes in common highly expressed and differentially expressed genes. RAP1 signaling pathway was used as the contact center and coordinated with other pathways to regulate placenta development. This study could provide insights into the molecular mechanisms underlying ruminant placentation. Abstract This study explored the trophoblast transcriptome to understand potential functional genes involved in early placental development in goats and their enriched signaling pathways. Trophoblast samples were collected from nine Dazu Black goats on days 20, 25, and 30 of pregnancy (D20, D25, and D30). As the pregnancy progressed, the morphology and histological structures showed significant growth, adhesion, and angiogenesis. A total of 23,253 commonly expressed genes (CEGs) and 4439 differently expressed genes (DEGs) were detected by RNA sequencing. The common highly expressed genes (ChEGs) (the top 100 CEGs) with the highest FPKM percentage (29.9%) of all CEGs were annotated to the ribosome pathway and maintain pregnancy. DEGs were abundant in D30 vs. D20 (3715 DEGs). Besides, the DEGs were associated with the inhibition of oxidative phosphorylation and activation of PI3K-Akt, focal adhesion, ECM–receptor interaction, Rap1, and CAM signaling pathways. The RAP1 may be a central pathway since it coordinates with others to regulate the cell proliferation, invasion, migration, and fusion of trophoblasts. qRT-PCR and Western blot analysis confirmed the transcriptional expression in IGF1, VEGFC, RAPGEF3, PIK3CA, AKT3, ITGB3, ITGA11, SPP1, NOS1, and ATP6V0B genes and protein levels in VEGF, RAPGEF3, and Akt. This is the first study of transcriptome profiling in goat placenta and provides diverse genetic resources for further research on placenta development.
Collapse
|
58
|
Chen H, Williams KE, Kwan EY, Kapidzic M, Puckett KA, Aburajab RK, Robinson JF, Fisher SJ. Global proteomic analyses of human cytotrophoblast differentiation/invasion. Development 2021; 148:dev199561. [PMID: 34121116 PMCID: PMC8276980 DOI: 10.1242/dev.199561] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/01/2021] [Indexed: 12/21/2022]
Abstract
During human pregnancy, cytotrophoblasts (CTBs) from the placenta differentiate into specialized subpopulations that play crucial roles in proper fetal growth and development. A subset of these CTBs differentiate along an invasive pathway, penetrating the decidua and anchoring the placenta to the uterus. A crucial hurdle in pregnancy is the ability of these cells to migrate, invade and remodel spiral arteries, ensuring adequate blood flow to nourish the developing fetus. Although advances continue in describing the molecular features regulating the differentiation of these cells, assessment of their global proteomic changes at mid-gestation remain undefined. Here, using sequential window acquisition of all theoretical fragment-ion spectra (SWATH), which is a data-independent acquisition strategy, we characterized the protein repertoire of second trimester human CTBs during their differentiation towards an invasive phenotype. This mass spectrometry-based approach allowed identification of 3026 proteins across four culture time points corresponding to sequential stages of differentiation, confirming the expression dynamics of established molecules and offering new information into other pathways involved. The availability of a SWATH CTB global spectral library serves as a beneficial resource for hypothesis generation and as a foundation for further understanding CTB differentiation dynamics.
Collapse
Affiliation(s)
- Hao Chen
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, CA 94143, USA
| | - Katherine E. Williams
- Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, CA 94143, USA
| | - Elaine Y. Kwan
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
| | - Mirhan Kapidzic
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
| | - Kenisha A. Puckett
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
| | - Rayyan K. Aburajab
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| | - Joshua F. Robinson
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
| | - Susan J. Fisher
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, CA 94143, USA
- Division of Maternal Fetal Medicine, University of California, San Francisco, CA 94143, USA
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
- Human Embryonic Stem Cell Program, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
59
|
Sanchez-Aranguren L, Nadeem S. Bioenergetics adaptations and redox homeostasis in pregnancy and related disorders. Mol Cell Biochem 2021; 476:4003-4018. [PMID: 34196872 PMCID: PMC8473347 DOI: 10.1007/s11010-021-04215-0] [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: 09/22/2020] [Accepted: 06/21/2021] [Indexed: 12/14/2022]
Abstract
Pregnancy is a challenging physiological process that involves maternal adaptations to the increasing energetics demands imposed by the growing conceptus. Failure to adapt to these requirements may result in serious health complications for the mother and the baby. The mitochondria are biosynthetic and energy-producing organelles supporting the augmented energetic demands of pregnancy. Evidence suggests that placental mitochondria display a dynamic phenotype through gestation. At early stages of pregnancy placental mitochondria are mainly responsible for the generation of metabolic intermediates and reactive oxygen species (ROS), while at later stages of gestation, the placental mitochondria exhibit high rates of oxygen consumption. This review describes the metabolic fingerprint of the placental mitochondria at different stages of pregnancy and summarises key signs of mitochondrial dysfunction in pathological pregnancy conditions, including preeclampsia, gestational diabetes and intrauterine growth restriction (IUGR). So far, the effects of placental-driven metabolic changes governing the metabolic adaptations occurring in different maternal tissues in both, healthy and pathological pregnancies, remain to be uncovered. Understanding the function and molecular aspects of the adaptations occurring in placental and maternal tissue's mitochondria will unveil potential targets for further therapeutic exploration that could address pregnancy-related disorders. Targeting mitochondrial metabolism is an emerging approach for regulating mitochondrial bioenergetics. This review will also describe the potential therapeutic use of compounds with a recognised effect on mitochondria, for the management of preeclampsia.
Collapse
Affiliation(s)
| | - Sarah Nadeem
- College of Health and Life Sciences, Aston Medical School, Aston University, Birmingham, UK
| |
Collapse
|
60
|
Chu T, Mouillet JF, Cao Z, Barak O, Ouyang Y, Sadovsky Y. RNA Network Interactions During Differentiation of Human Trophoblasts. Front Cell Dev Biol 2021; 9:677981. [PMID: 34150771 PMCID: PMC8209545 DOI: 10.3389/fcell.2021.677981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
In the human placenta, two trophoblast cell layers separate the maternal blood from the villous basement membrane and fetal capillary endothelial cells. The inner layer, which is complete early in pregnancy and later becomes discontinuous, comprises the proliferative mononuclear cytotrophoblasts, which fuse together and differentiate to form the outer layer of multinucleated syncytiotrophoblasts. Because the syncytiotrophoblasts are responsible for key maternal-fetal exchange functions, tight regulation of this differentiation process is critical for the proper development and the functional role of the placenta. The molecular mechanisms regulating the fusion and differentiation of trophoblasts during human pregnancy remain poorly understood. To decipher the interactions of non-coding RNAs (ncRNAs) in this process, we exposed cultured primary human trophoblasts to standard in vitro differentiation conditions or to conditions known to hinder this differentiation process, namely exposure to hypoxia (O2 < 1%) or to the addition of dimethyl sulfoxide (DMSO, 1.5%) to the culture medium. Using next generation sequencing technology, we analyzed the differential expression of trophoblastic lncRNAs, miRNAs, and mRNAs that are concordantly modulated by both hypoxia and DMSO. Additionally, we developed a model to construct a lncRNA-miRNA-mRNA co-expression network and inferred the functions of lncRNAs and miRNAs via indirect gene ontology analysis. This study improves our knowledge of the interactions between ncRNAs and mRNAs during trophoblast differentiation and identifies key biological processes that may be impaired in common gestational diseases, such as fetal growth restriction or preeclampsia.
Collapse
Affiliation(s)
- Tianjiao Chu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jean-Francois Mouillet
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Zhishen Cao
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Oren Barak
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Yingshi Ouyang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Yoel Sadovsky
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
61
|
Varberg KM, Soares MJ. Paradigms for investigating invasive trophoblast cell development and contributions to uterine spiral artery remodeling. Placenta 2021; 113:48-56. [PMID: 33985793 DOI: 10.1016/j.placenta.2021.04.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 04/16/2021] [Accepted: 04/18/2021] [Indexed: 12/21/2022]
Abstract
Uterine spiral arteries are extensively remodeled during placentation to ensure sufficient delivery of maternal blood to the developing fetus. Uterine spiral arterial remodeling is complex, as cells originating from both mother and developing conceptus interact at the maternal interface to regulate the extracellular matrix remodeling and vasculature restructuring necessary for successful placentation. Despite this complexity, one mechanism critical to spiral artery remodeling is trophoblast cell invasion into the maternal compartment. Invasive trophoblast cells include both interstitial and endovascular populations that exhibit spatiotemporal differences in uterine invasion, including proximity to uterine spiral arteries. Interstitial trophoblast cells invade the uterine parenchyma where they are interspersed among stromal cells. Endovascular trophoblast cells infiltrate uterine spiral arteries, replace endothelial cells, adopt a pseudo-endothelial cell phenotype, and engineer vessel remodeling. Impaired trophoblast cell invasion and, consequently, insufficient uterine spiral arterial remodeling can lead to the development of pregnancy disorders, such as preeclampsia, intrauterine growth restriction, and premature birth. This review provides insights into invasive trophoblast cells and their function during normal placentation as well as in settings of disease.
Collapse
Affiliation(s)
- Kaela M Varberg
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA.
| | - Michael J Soares
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA; Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Center for Perinatal Research, Children's Mercy Research Institute, Children's Mercy Kansas City, Missouri 64108, USA.
| |
Collapse
|
62
|
Modeling preeclampsia using human induced pluripotent stem cells. Sci Rep 2021; 11:5877. [PMID: 33723311 PMCID: PMC7961010 DOI: 10.1038/s41598-021-85230-5] [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: 11/18/2020] [Accepted: 02/26/2021] [Indexed: 12/15/2022] Open
Abstract
Preeclampsia (PE) is a pregnancy-specific hypertensive disorder, affecting up to 10% of pregnancies worldwide. The primary etiology is considered to be abnormal development and function of placental cells called trophoblasts. We previously developed a two-step protocol for differentiation of human pluripotent stem cells, first into cytotrophoblast (CTB) progenitor-like cells, and then into both syncytiotrophoblast (STB)- and extravillous trophoblast (EVT)-like cells, and showed that it can model both normal and abnormal trophoblast differentiation. We have now applied this protocol to induced pluripotent stem cells (iPSC) derived from placentas of pregnancies with or without PE. While there were no differences in CTB induction or EVT formation, PE-iPSC-derived trophoblast showed a defect in syncytialization, as well as a blunted response to hypoxia. RNAseq analysis showed defects in STB formation and response to hypoxia; however, DNA methylation changes were minimal, corresponding only to changes in response to hypoxia. Overall, PE-iPSC recapitulated multiple defects associated with placental dysfunction, including a lack of response to decreased oxygen tension. This emphasizes the importance of the maternal microenvironment in normal placentation, and highlights potential pathways that can be targeted for diagnosis or therapy, while absence of marked DNA methylation changes suggests that other regulatory mechanisms mediate these alterations.
Collapse
|
63
|
Molecular and immunological developments in placentas. Hum Immunol 2021; 82:317-324. [PMID: 33581928 DOI: 10.1016/j.humimm.2021.01.012] [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: 11/27/2020] [Revised: 01/03/2021] [Accepted: 01/21/2021] [Indexed: 12/20/2022]
Abstract
Cytotrophoblasts differentiate in two directions during early placentation: syncytiotrophoblasts (STBs) and extravillous trophoblasts (EVTs). STBs face maternal immune cells in placentas, and EVTs, which invade the decidua and uterine myometrium, face the cells in the uterus. This situation, in which trophoblasts come into contact with maternal immune cells, is known as the maternal-fetal interface. Despite fetuses and fetus-derived trophoblast cells being of the semi-allogeneic conceptus, fetuses and placentas are not rejected by the maternal immune system because of maternal-fetal tolerance. The acquired tolerance develops during normal placentation, resulting in normal fetal development in humans. In this review, we introduce placental development from the viewpoint of molecular biology. In addition, we discuss how the disruption of placental development could lead to complications in pregnancy, such as hypertensive disorder of pregnancy, fetal growth restriction, or miscarriage.
Collapse
|
64
|
Baryla I, Pluciennik E, Kośla K, Wojcik M, Zieleniak A, Zurawska-Klis M, Cypryk K, Wozniak LA, Bednarek AK. Identification of a novel association for the WWOX/HIF1A axis with gestational diabetes mellitus (GDM). PeerJ 2021; 9:e10604. [PMID: 33520443 PMCID: PMC7811782 DOI: 10.7717/peerj.10604] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/28/2020] [Indexed: 12/12/2022] Open
Abstract
Background Although the WW-domain-containing oxidoreductase (WWOX)/Hypoxia-inducible factor 1 (HIF1) pathway is a well-known regulator of cellular glucose and energy metabolism in pathophysiological processes, its role in gestational diabetes mellitus (GDM), remains elusive. We undertook this study to determine the effect of WWOX/HIF1A signaling on the expression of glucose metabolism genes in GDM patients. Methods Leukocytes were obtained from 135 pregnant women with (n = 98) or without (n = 37) GDM and, in turn, 3 months (n = 8) and 1 year (n = 12) postpartum. Quantitative RT-PCR was performed to determine gene expression profiles of the WWOX/HIF1A-related genes, including those involved in glucose transport (SLC2A1, SLC2A4), glycolytic pathway (HK2, PKM2, PFK, LDHA), Wnt pathway (DVL2, CTNNB1), and inflammatory response (NFKB1). Results GDM patients displayed a significant downregulation of WWOX with simultaneous upregulation of HIF1A which resulted in approximately six times reduction in WWOX/HIF1A ratio. As a consequence, HIF1A induced genes (SLC2A1, HK2, PFK, PKM) were found to be overexpressed in GDM compared to normal pregnancy and negative correlate with WWOX/HIF1A ratio. The postpartum WWOX expression was higher than during GDM, but its level was comparable to that observed in normal pregnancy. Conclusions The obtained results suggest a significant contribution of the WWOX gene to glucose metabolism in patients with gestational diabetes. Decreased WWOX expression in GDM compared to normal pregnancy, and in particular reduction of WWOX/HIF1A ratio, indicate that WWOX modulates HIF1α activity in normal tissues as described in the tumor. The effect of HIF1α excessive activation is to increase the expression of genes encoding proteins directly involved in the glycolysis which may lead to pathological changes in glucose metabolism observed in gestational diabetes.
Collapse
Affiliation(s)
- Izabela Baryla
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Elzbieta Pluciennik
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Katarzyna Kośla
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Marzena Wojcik
- Department of Structural Biology, Medical University of Lodz, Lodz, Poland
| | - Andrzej Zieleniak
- Department of Structural Biology, Medical University of Lodz, Lodz, Poland
| | - Monika Zurawska-Klis
- Department of Internal Diseases and Diabetology, Medical University of Lodz, Lodz, Poland
| | - Katarzyna Cypryk
- Department of Internal Diseases and Diabetology, Medical University of Lodz, Lodz, Poland
| | | | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
65
|
Chen C, Kang X, Li C, Guo F, Wang Q, Zhao A. Involvement of signal transducers and activators of transcription in trophoblast differentiation. Placenta 2021; 105:94-103. [PMID: 33556719 DOI: 10.1016/j.placenta.2021.01.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/27/2020] [Accepted: 01/27/2021] [Indexed: 12/21/2022]
Abstract
INTRODUCTION To explore the involvement of signal transducers and activators of transcription (STATs) in trophoblast differentiation. METHODS AND RESULTS First, the localization of STATs in human placentas was detected via immunohistochemistry (IHC) and immunofluorescence (IF). Cytotrophoblasts (CTBs) expressed both STAT1 and 3, but syncytiotrophoblasts (STBs) did not. Staining for these two proteins showed a distinct upregulation from the proximal part to the distal end of cell columns. STAT5B was mainly expressed in the STBs, low in the CTBs, and absent in the extravillous trophoblasts (EVTs). Next, the 44 placenta samples were tested via western blot (WB) and quantitative real time polymerase chain reaction (qRT-PCR). We found a decrease in STAT1 and 3 and an increase in STAT5B as gestation increased from five to 10 weeks. Then, an in vitro co-culture model of placenta with or without decidua stromal cells (DSCs), as detected via flow cytometry, revealed an increase in the human leukocyte antigen (HLA)-G positive rate in trophoblasts from placentas co-cultured with DSCs, accompanied by an increase in p-STAT1 and 3 and a decrease in p-STAT5 and STAT5B. Finally, mRNA of matrix metalloproteinases (MMPs) and integrins after STAT silencing in HTR-8/SVneo was detected via qRT-PCR. STAT1 silencing decreased MMP9 expression, STAT3 silencing decreased MMP9, integrin α6, and β4 expression, and STAT5B silencing increased MMP2 and integrin β1 expression. DISCUSSION Different trophoblasts showed distinct STAT expression profiles which were related to their MMP and integrin expression. DSCs promoted trophoblast differentiation into EVTs, possibly by regulating the STAT expression of the trophoblasts.
Collapse
Affiliation(s)
- Chao Chen
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China
| | - Xiaomin Kang
- Department of Reproductive Medical Center, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Congcong Li
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China
| | - Feng Guo
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China
| | - Qiaohong Wang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China
| | - Aimin Zhao
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China.
| |
Collapse
|
66
|
Yuan D, Yang Z, Chen Y, Li S, Tan B, Yu Q. Hypoxia-induced SPOP attenuates the mobility of trophoblast cells through inhibition of the PI3K/AKT/GSK3β pathway. Cell Biol Int 2021; 45:599-611. [PMID: 33200474 DOI: 10.1002/cbin.11501] [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: 09/01/2020] [Revised: 10/20/2020] [Accepted: 11/11/2020] [Indexed: 12/19/2022]
Abstract
Placental hypoxia has been implicated in pregnancy pathologies such as pre-eclampsia and intrauterine growth restriction. However, the underlying mechanism by which the trophoblasts respond to hypoxia remains unclear. Speckle-type POZ protein (SPOP), an E3 ubiquitin ligase adapter, was previously reported to play important roles in various physiological and pathological processes. This study aims to investigate the expression and biological functions of SPOP after exposure to cobalt chloride (CoCl2 )-mimicked hypoxia conditions using human trophoblast-derived choriocarcinoma cell lines and extravillous cytotrophoblast. These data showed that SPOP protein was directly induced by CoCl2 -mimicked hypoxia and regulated by HIF-1α at the posttranscription level. CoCl2 treatment could dramatically influence the localization of SPOP in trophoblasts, especially the accumulation of SPOP into the nucleus. In addition, both CoCl2 -mimicked hypoxia and induction of endogenous SPOP expression by lentivirus transfection attenuated the migration and invasion abilities of trophoblasts. Furthermore, we demonstrated that SPOP was involved in CoCl2 -induced the inhibition of the PI3K/AKT/GSK3β pathway in placental trophoblasts. Taken together, these data indicate that accumulation of HIF-1α augments the expression of SPOP in trophoblasts, which impairs trophoblastic mobility by targeting the PI3K/AKT/GSK3β pathway. This potentially leads to insufficient uterine spiral artery remodeling and suboptimal placental perfusion, and thus the development of pregnancy-related complication.
Collapse
Affiliation(s)
- Dong Yuan
- Department of Gynecology, Chongqing Medical University Affiliated Second Hospital, Chongqing, China
| | - Zhu Yang
- Department of Gynecology, Chongqing Medical University Affiliated Second Hospital, Chongqing, China.,Molecular Medical Laboratory, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Yiyu Chen
- Department of Clinical Laboratory, Chongqing Medical University Affiliated Stomatological Hospital, Chongqing, China
| | - Siyuan Li
- Department of Laboratory Medicine, Chongqing Medical University Affiliated Children's Hospital, Chongqing, China
| | - Benxu Tan
- Department of Oncology, Chongqing Medical University Affiliated First Hospital, Chongqing, China
| | - Qiubo Yu
- Molecular Medical Laboratory, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| |
Collapse
|
67
|
Role of Peroxisome Proliferator-Activated Receptors (PPARs) in Trophoblast Functions. Int J Mol Sci 2021; 22:ijms22010433. [PMID: 33406768 PMCID: PMC7795665 DOI: 10.3390/ijms22010433] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 12/23/2020] [Accepted: 12/29/2020] [Indexed: 12/11/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARα, PPARβ/δ, and PPARγ) belong to the transcription factor family, and they are highly expressed in all types of trophoblast during pregnancy. The present review discusses currently published papers that are related to the regulation of PPARs via lipid metabolism, glucose metabolism, and amino acid metabolism to affect trophoblast physiological conditions, including differentiation, maturation, secretion, fusion, proliferation, migration, and invasion. Recent pieces of evidence have proven that the dysfunctions of PPARs in trophoblast lead to several related pregnancy diseases such as recurrent miscarriage, preeclampsia, intrauterine growth restriction, and gestational diabetes mellitus. Moreover, the underlying mechanisms of PPARs in the control of these processes have been discussed as well. Finally, this review's purposes are to provide more knowledge about the role of PPARs in normal and disturbed pregnancy with trophoblast, so as to find PPAR ligands as a potential therapeutic target in the treatment and prevention of adverse pregnancy outcomes.
Collapse
|
68
|
Colson A, Sonveaux P, Debiève F, Sferruzzi-Perri AN. Adaptations of the human placenta to hypoxia: opportunities for interventions in fetal growth restriction. Hum Reprod Update 2020; 27:531-569. [PMID: 33377492 DOI: 10.1093/humupd/dmaa053] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/15/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The placenta is the functional interface between the mother and the fetus during pregnancy, and a critical determinant of fetal growth and life-long health. In the first trimester, it develops under a low-oxygen environment, which is essential for the conceptus who has little defense against reactive oxygen species produced during oxidative metabolism. However, failure of invasive trophoblasts to sufficiently remodel uterine arteries toward dilated vessels by the end of the first trimester can lead to reduced/intermittent blood flow, persistent hypoxia and oxidative stress in the placenta with consequences for fetal growth. Fetal growth restriction (FGR) is observed in ∼10% of pregnancies and is frequently seen in association with other pregnancy complications, such as preeclampsia (PE). FGR is one of the main challenges for obstetricians and pediatricians, as smaller fetuses have greater perinatal risks of morbidity and mortality and postnatal risks of neurodevelopmental and cardio-metabolic disorders. OBJECTIVE AND RATIONALE The aim of this review was to examine the importance of placental responses to changing oxygen environments during abnormal pregnancy in terms of cellular, molecular and functional changes in order to highlight new therapeutic pathways, and to pinpoint approaches aimed at enhancing oxygen supply and/or mitigating oxidative stress in the placenta as a mean of optimizing fetal growth. SEARCH METHODS An extensive online search of peer-reviewed articles using PubMed was performed with combinations of search terms including pregnancy, placenta, trophoblast, oxygen, hypoxia, high altitude, FGR and PE (last updated in May 2020). OUTCOMES Trophoblast differentiation and placental establishment are governed by oxygen availability/hypoxia in early pregnancy. The placental response to late gestational hypoxia includes changes in syncytialization, mitochondrial functions, endoplasmic reticulum stress, hormone production, nutrient handling and angiogenic factor secretion. The nature of these changes depends on the extent of hypoxia, with some responses appearing adaptive and others appearing detrimental to the placental support of fetal growth. Emerging approaches that aim to increase placental oxygen supply and/or reduce the impacts of excessive oxidative stress are promising for their potential to prevent/treat FGR. WIDER IMPLICATIONS There are many risks and challenges of intervening during pregnancy that must be considered. The establishment of human trophoblast stem cell lines and organoids will allow further mechanistic studies of the effects of hypoxia and may lead to advanced screening of drugs for use in pregnancies complicated by placental insufficiency/hypoxia. Since no treatments are currently available, a better understanding of placental adaptations to hypoxia would help to develop therapies or repurpose drugs to optimize placental function and fetal growth, with life-long benefits to human health.
Collapse
Affiliation(s)
- Arthur Colson
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Frédéric Debiève
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
69
|
Io S, Kondoh E, Chigusa Y, Kawasaki K, Mandai M, Yamada AS. New era of trophoblast research: integrating morphological and molecular approaches. Hum Reprod Update 2020; 26:611-633. [PMID: 32728695 DOI: 10.1093/humupd/dmaa020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/24/2020] [Accepted: 04/20/2020] [Indexed: 12/27/2022] Open
Abstract
Many pregnancy complications are the result of dysfunction in the placenta. The pathogenic mechanisms of placenta-mediated pregnancy complications, however, are unclear. Abnormal placental development in these conditions begins in the first trimester, but no symptoms are observed during this period. To elucidate effective preventative treatments, understanding the differentiation and development of human placenta is crucial. This review elucidates the uniqueness of the human placenta in early development from the aspect of structural characteristics and molecular markers. We summarise the morphogenesis of human placenta based on human specimens and then compile molecular markers that have been clarified by immunostaining and RNA-sequencing data across species. Relevant studies were identified using the PubMed database and Google Scholar search engines up to March 2020. All articles were independently screened for eligibility by the authors based on titles and abstracts. In particular, the authors carefully examined literature on human placentation. This review integrates the development of human placentation from morphological approaches in comparison with other species and provides new insights into trophoblast molecular markers. The morphological features of human early placentation are described in Carnegie stages (CS), from CS3 (floating blastocyst) to CS9 (emerging point of tertiary villi). Molecular markers are described for each type of trophoblast involved in human placental development. We summarise the character of human trophoblast cell lines and explain how long-term culture system of human cytotrophoblast, both monolayer and spheroid, established in recent studies allows for the generation of human trophoblast cell lines. Due to differences in developmental features among species, it is desirable to understand early placentation in humans. In addition, reliable molecular markers that reflect normal human trophoblast are needed to advance trophoblast research. In the clinical setting, these markers can be valuable means for morphologically and functionally assessing placenta-mediated pregnancy complications and provide early prediction and management of these diseases.
Collapse
Affiliation(s)
- Shingo Io
- Department of Life Science Frontiers, Center for iPS Cell Research & Application, Kyoto University, Kyoto, Japan.,Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan
| | - Eiji Kondoh
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshitsugu Chigusa
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kaoru Kawasaki
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - And Shigehito Yamada
- Human Health Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Congenital Anomaly Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
70
|
Adu-Gyamfi EA, Czika A, Gorleku PN, Ullah A, Panhwar Z, Ruan LL, Ding YB, Wang YX. The Involvement of Cell Adhesion Molecules, Tight Junctions, and Gap Junctions in Human Placentation. Reprod Sci 2020; 28:305-320. [PMID: 33146876 DOI: 10.1007/s43032-020-00364-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023]
Abstract
Placentation is a major determinant of the success of pregnancy. It is regulated by several factors such as cell adhesion molecules, tight junctions, and gap junctions. The cell adhesion molecules are integrins, cadherins, immunoglobulins, nectins, and selectins. The tight junctions are composed of claudins, occludin, and junction adhesion molecule proteins while the gap junctions are composed of connexins of varying molecular weights. During placentation, some of these molecules regulate trophoblast proliferation, trophoblast fusion, trophoblast migration, trophoblast invasion, trophoblast-endothelium adhesion, glandular remodeling, and spiral artery remodeling. There is a dysregulated placental expression of some of these molecules during obstetric complications. We have, hereby, indicated the expression patterns of the subunits of each of these molecules in the various trophoblast subtypes and in the decidua, and have highlighted their involvement in physiological and pathological placentation. The available evidence points to the relevance of these molecules as distinguishing markers of the various trophoblast lineages and as potential therapeutic targets in the management of malplacentation-mediated diseases.
Collapse
Affiliation(s)
- Enoch Appiah Adu-Gyamfi
- Department of Reproductive Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Box 197, No. 1 Yixueyuan Rd, Chongqing, 400016, People's Republic of China.
| | - Armin Czika
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Box 197, No. 1 Yixueyuan Rd, Chongqing, 400016, People's Republic of China
| | - Philip Narteh Gorleku
- Department of Medical Imaging, School of Medical Sciences, University of Cape Coast, Cape Coast, Republic of Ghana
| | - Amin Ullah
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Box 197, No. 1 Yixueyuan Rd, Chongqing, 400016, People's Republic of China
| | - Zulqarnain Panhwar
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Box 197, No. 1 Yixueyuan Rd, Chongqing, 400016, People's Republic of China
| | - Ling-Ling Ruan
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Box 197, No. 1 Yixueyuan Rd, Chongqing, 400016, People's Republic of China
| | - Yu-Bin Ding
- Department of Reproductive Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Box 197, No. 1 Yixueyuan Rd, Chongqing, 400016, People's Republic of China.
| | - Ying-Xiong Wang
- Department of Reproductive Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Box 197, No. 1 Yixueyuan Rd, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
71
|
Castel G, Meistermann D, Bretin B, Firmin J, Blin J, Loubersac S, Bruneau A, Chevolleau S, Kilens S, Chariau C, Gaignerie A, Francheteau Q, Kagawa H, Charpentier E, Flippe L, François-Campion V, Haider S, Dietrich B, Knöfler M, Arima T, Bourdon J, Rivron N, Masson D, Fournier T, Okae H, Fréour T, David L. Induction of Human Trophoblast Stem Cells from Somatic Cells and Pluripotent Stem Cells. Cell Rep 2020; 33:108419. [PMID: 33238118 DOI: 10.1016/j.celrep.2020.108419] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 08/21/2020] [Accepted: 10/29/2020] [Indexed: 12/31/2022] Open
Abstract
Human trophoblast stem cells (hTSCs) derived from blastocysts and first-trimester cytotrophoblasts offer an unprecedented opportunity to study the placenta. However, access to human embryos and first-trimester placentas is limited, thus preventing the establishment of hTSCs from diverse genetic backgrounds associated with placental disorders. Here, we show that hTSCs can be generated from numerous genetic backgrounds using post-natal cells via two alternative methods: (1) somatic cell reprogramming of adult fibroblasts with OCT4, SOX2, KLF4, MYC (OSKM) and (2) cell fate conversion of naive and extended pluripotent stem cells. The resulting induced/converted hTSCs recapitulated hallmarks of hTSCs including long-term self-renewal, expression of specific transcription factors, transcriptomic signature, and the potential to differentiate into syncytiotrophoblast and extravillous trophoblast cells. We also clarified the developmental stage of hTSCs and show that these cells resemble day 8 cytotrophoblasts. Altogether, hTSC lines of diverse genetic origins open the possibility to model both placental development and diseases in a dish.
Collapse
Affiliation(s)
- Gaël Castel
- Université de Nantes, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44000 Nantes, France
| | - Dimitri Meistermann
- Université de Nantes, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44000 Nantes, France; LS2N, Université de Nantes, CNRS, Nantes, France
| | - Betty Bretin
- Université de Nantes, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44000 Nantes, France
| | - Julie Firmin
- Université de Nantes, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44000 Nantes, France; Service de Biologie de la Reproduction, CHU Nantes, Nantes, France
| | - Justine Blin
- CHU Nantes, Laboratory of Clinical Biochemistry, Nantes, France
| | - Sophie Loubersac
- Service de Biologie de la Reproduction, CHU Nantes, Nantes, France
| | - Alexandre Bruneau
- Université de Nantes, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44000 Nantes, France
| | - Simon Chevolleau
- Université de Nantes, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44000 Nantes, France
| | - Stéphanie Kilens
- Université de Nantes, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44000 Nantes, France
| | - Caroline Chariau
- Université de Nantes, CHU Nantes, SFR Santé, FED 4203, INSERM UMS 016, CNRS UMS 3556, Nantes, France
| | - Anne Gaignerie
- Université de Nantes, CHU Nantes, SFR Santé, FED 4203, INSERM UMS 016, CNRS UMS 3556, Nantes, France
| | - Quentin Francheteau
- Université de Nantes, CHU Nantes, SFR Santé, FED 4203, INSERM UMS 016, CNRS UMS 3556, Nantes, France
| | - Harunobu Kagawa
- Institute of Molecular Biotechnology, Austrian Academy of Science, Vienna, Austria
| | - Eric Charpentier
- Université de Nantes, CHU Nantes, SFR Santé, FED 4203, INSERM UMS 016, CNRS UMS 3556, Nantes, France
| | - Léa Flippe
- Université de Nantes, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44000 Nantes, France
| | - Valentin François-Campion
- Université de Nantes, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44000 Nantes, France
| | - Sandra Haider
- Department of Obstetrics and Gynaecology, Medical University of Vienna, Reproductive Biology Unit, Währinger Gürtel 18-20, 5Q, 1090 Vienna, Austria
| | - Bianca Dietrich
- Department of Obstetrics and Gynaecology, Medical University of Vienna, Reproductive Biology Unit, Währinger Gürtel 18-20, 5Q, 1090 Vienna, Austria
| | - Martin Knöfler
- Department of Obstetrics and Gynaecology, Medical University of Vienna, Reproductive Biology Unit, Währinger Gürtel 18-20, 5Q, 1090 Vienna, Austria
| | - Takahiro Arima
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | | | - Nicolas Rivron
- Institute of Molecular Biotechnology, Austrian Academy of Science, Vienna, Austria
| | - Damien Masson
- CHU Nantes, Laboratory of Clinical Biochemistry, Nantes, France; Université de Nantes, INSERM, U1235, Nantes, France
| | - Thierry Fournier
- Université de Paris, INSERM, UMR-S 1139, 3PHM, 75006 Paris, France
| | - Hiroaki Okae
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Thomas Fréour
- Université de Nantes, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44000 Nantes, France; Service de Biologie de la Reproduction, CHU Nantes, Nantes, France
| | - Laurent David
- Université de Nantes, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44000 Nantes, France; Université de Nantes, CHU Nantes, SFR Santé, FED 4203, INSERM UMS 016, CNRS UMS 3556, Nantes, France.
| |
Collapse
|
72
|
Bao H, Liu D, Xu Y, Sun Y, Mu C, Yu Y, Wang C, Han Q, Liu S, Cai H, Liu F, Kong S, Deng W, Cao B, Wang H, Wang Q, Lu J. Hyperactivated Wnt-β-catenin signaling in the absence of sFRP1 and sFRP5 disrupts trophoblast differentiation through repression of Ascl2. BMC Biol 2020; 18:151. [PMID: 33109217 PMCID: PMC7592576 DOI: 10.1186/s12915-020-00883-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 09/29/2020] [Indexed: 01/04/2023] Open
Abstract
Background Wnt signaling is a critical determinant for the maintenance and differentiation of stem/progenitor cells, including trophoblast stem cells during placental development. Hyperactivation of Wnt signaling has been shown to be associated with human trophoblast diseases. However, little is known about the impact and underlying mechanisms of excessive Wnt signaling during placental trophoblast development. Results In the present work, we observed that two inhibitors of Wnt signaling, secreted frizzled-related proteins 1 and 5 (Sfrp1 and Sfrp5), are highly expressed in the extraembryonic trophoblast suggesting possible roles in early placental development. Sfrp1 and Sfrp5 double knockout mice exhibited disturbed trophoblast differentiation in the placental ectoplacental cone (EPC), which contains the precursors of trophoblast giant cells (TGCs) and spongiotrophoblast cells. In addition, we employed mouse models expressing a truncated β-catenin with exon 3 deletion globally and trophoblast-specifically, as well as trophoblast stem cell lines, and unraveled that hyperactivation of canonical Wnt pathway exhausted the trophoblast precursor cells in the EPC, resulting in the overabundance of giant cells at the expense of spongiotrophoblast cells. Further examination uncovered that hyperactivation of canonical Wnt pathway disturbed trophoblast differentiation in the EPC via repressing Ascl2 expression. Conclusions Our investigations provide new insights that the homeostasis of canonical Wnt-β-catenin signaling is essential for EPC trophoblast differentiation during placental development, which is of high clinical relevance, since aberrant Wnt signaling is often associated with trophoblast-related diseases.
Collapse
Affiliation(s)
- Haili Bao
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, Fujian, People's Republic of China.,Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
| | - Dong Liu
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Yingchun Xu
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Yang Sun
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Change Mu
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Yongqin Yu
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Chunping Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Qian Han
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Sanmei Liu
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Han Cai
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, Fujian, People's Republic of China.,Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Fan Liu
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Shuangbo Kong
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, Fujian, People's Republic of China.,Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Wenbo Deng
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, Fujian, People's Republic of China.,Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Bin Cao
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, Fujian, People's Republic of China.,Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China
| | - Haibin Wang
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, Fujian, People's Republic of China. .,Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China.
| | - Qiang Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China. .,Department of Surgery, The Ohio State University Wexner Medical Center, Ohio, 43210, Columbus, USA.
| | - Jinhua Lu
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, Fujian, People's Republic of China. .,Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China.
| |
Collapse
|
73
|
Colson A, Depoix CL, Baldin P, Hubinont C, Sonveaux P, Debiève F. Hypoxia-inducible factor 2 alpha impairs human cytotrophoblast syncytialization: New insights into placental dysfunction and fetal growth restriction. FASEB J 2020; 34:15222-15235. [PMID: 32954526 DOI: 10.1096/fj.202001681r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/26/2020] [Accepted: 09/03/2020] [Indexed: 01/01/2023]
Abstract
Insufficient remodeling of uterine arteries causes pregnancy-related diseases, including fetal growth restriction and preeclampsia. In these situations, reduced maternal blood flow in the placenta is thought to be responsible for the persistence of a low oxygen environment throughout pregnancy. We hypothesized that chronic activation of transcription factors hypoxia-inducible factors (HIFs) actively participates in placental underdevelopment, which impairs fetal growth. The computer-assisted analysis in pathological placentas revealed an increased number of HIF-2α-positive nuclei in the syncytium compared to normal human placentas, while HIF-1α stabilization was unchanged. Specific involvement of HIF-2α was confirmed in primary human cytotrophoblasts rendered deficient for HIF1A or HIF2A. Silencing HIF2A increased the expression of main syncytialization markers as well as differentiation and syncytium formation. It also improved placental growth factor bioavailability. None of these changes was seen when silencing HIF1A. Conversely, the experimental induction of HIF-2α expression repressed forskolin-induced differentiation in BeWo choriocarcinoma cells. Our mechanistic insights evidence that transcription factor HIF-2α impairs placental function, thus suggesting its participation in fetal growth restriction and preeclampsia when placentas become chronically hypoxic. Furthermore, it suggests the possibility to develop novel molecular targeting therapies for placental dysfunction.
Collapse
Affiliation(s)
- Arthur Colson
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium.,Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Christophe Louis Depoix
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Pamela Baldin
- Department of Pathology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Corinne Hubinont
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium.,Department of Obstetrics, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Frédéric Debiève
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium.,Department of Obstetrics, Cliniques universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
74
|
Horii M, Bui T, Touma O, Cho HY, Parast MM. An Improved Two-Step Protocol for Trophoblast Differentiation of Human Pluripotent Stem Cells. ACTA ACUST UNITED AC 2020; 50:e96. [PMID: 31479595 DOI: 10.1002/cpsc.96] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We previously established a two-step protocol for differentiation of human pluripotent stem cells (hPSCs) into trophoblasts, using a StemPro-based minimal medium (EMIM) with bone morphogenetic protein-4 (BMP4). This protocol was suboptimal, resulting in induction of mixed mesoderm and trophoblast markers. Furthermore, adapting hPSCs to StemPro has proven difficult, and prolonged culture in this medium has been shown to promote genomic instability. Therefore, we moved on to the use of new media, including E8, and most recently, StemFlex, for rapid adaptation from feeder to non-feeder conditions. In the new protocol, we have incorporated the WNT inhibitor IWP2 into the first step, resulting in uniform differentiation of hPSCs into cytotrophoblast (CTB)-like cells, without induction of the mesoderm lineage. We also show that, at the end of the second step, there are distinct populations of terminally differentiated multinucleated human chorionic gonadotropin (hCG)-producing syncytiotrophoblast (STB) and HLAG+ extravillous trophoblast (EVT)-like cells. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Mariko Horii
- Department of Pathology, University of California San Diego, La Jolla, California.,Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Tony Bui
- Department of Pathology, University of California San Diego, La Jolla, California.,Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Ojeni Touma
- Department of Pathology, University of California San Diego, La Jolla, California.,Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Hee Young Cho
- Department of Pathology, University of California San Diego, La Jolla, California.,Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, California.,Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| |
Collapse
|
75
|
Role of Hippo signaling pathway in early placental development. Proc Natl Acad Sci U S A 2020; 117:20354-20356. [PMID: 32788372 DOI: 10.1073/pnas.2013559117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
76
|
Placental function in maternal obesity. Clin Sci (Lond) 2020; 134:961-984. [PMID: 32313958 DOI: 10.1042/cs20190266] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023]
Abstract
Maternal obesity is associated with pregnancy complications and increases the risk for the infant to develop obesity, diabetes and cardiovascular disease later in life. However, the mechanisms linking the maternal obesogenic environment to adverse short- and long-term outcomes remain poorly understood. As compared with pregnant women with normal BMI, women entering pregnancy obese have more pronounced insulin resistance, higher circulating plasma insulin, leptin, IGF-1, lipids and possibly proinflammatory cytokines and lower plasma adiponectin. Importantly, the changes in maternal levels of nutrients, growth factors and hormones in maternal obesity modulate placental function. For example, high insulin, leptin, IGF-1 and low adiponectin in obese pregnant women activate mTOR signaling in the placenta, promoting protein synthesis, mitochondrial function and nutrient transport. These changes are believed to increase fetal nutrient supply and contribute to fetal overgrowth and/or adiposity in offspring, which increases the risk to develop disease later in life. However, the majority of obese women give birth to normal weight infants and these pregnancies are also associated with activation of inflammatory signaling pathways, oxidative stress, decreased oxidative phosphorylation and lipid accumulation in the placenta. Recent bioinformatics approaches have expanded our understanding of how maternal obesity affects the placenta; however, the link between changes in placental function and adverse outcomes in obese women giving birth to normal sized infants is unclear. Interventions that specifically target placental function, such as activation of placental adiponectin receptors, may prevent the transmission of metabolic disease from obese women to the next generation.
Collapse
|
77
|
Decidual Vasculopathy and Spiral Artery Remodeling Revisited III: Hypoxia and Re-oxygenation Sequence with Vascular Regeneration. REPRODUCTIVE MEDICINE 2020. [DOI: 10.3390/reprodmed1020006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Aim: Spiral artery remodeling at early pregnancy is characterized by two distinct mechanisms with two morphologic features, namely, trophoblastic-dependent vascular invasion with “plugging”, and trophoblastic-independent mural muscular hypertrophy/hyperplasia, both of which lead to the blocking or narrowing of the arterial lumen with the consequence of reduced maternal blood flow to the developing embryo. Methods: Review of historic literature in light of the new discovery of CD56 (NCAM) expression on endovascular trophoblasts at late gestation, in relation to placental lateral growth with vascular regeneration. Results: Reduced maternal blood flow to the embryo results in a hypoxic condition critical for trophectoderm differentiation and proliferation. Hypoxia is also important for the development of hemangioblasts of vasculogenesis, and hematopoiesis of the placental villi. Up to 13 weeks, both uteroplacental and fetoplacental circulations are established and hypoxic condition relieved for normal fetal/placenta development by ultrasonography. The persistence of trophoblastic plugging and/or mural muscular hypertrophy/hyperplasia leads to persistent reduced maternal blood flow to the placenta, resulting in persistent hypoxia and increased angiogenesis, with a constellation of pathologic features of maternal vascular malperfusion atlate gestation. Wilm’s tumor gene (WT1) expression appears to be central to steroid and peptide hormonal actions in early pregnancy, and vascular regeneration/restoration after pregnancy. Conclusions: Spiral artery remodeling at early pregnancy leads to hypoxia with vascular transformation, and the establishment of uteroplacental circulation results in relief of hypoxia. The hypoxia–re-oxygenation sequence may provide insights into the mechanism of normal fetal/placental development and associated pregnancy complications, such as preeclampsia.
Collapse
|
78
|
Horii M, Touma O, Bui T, Parast MM. Modeling human trophoblast, the placental epithelium at the maternal fetal interface. Reproduction 2020; 160:R1-R11. [PMID: 32485667 PMCID: PMC7286067 DOI: 10.1530/rep-19-0428] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 04/17/2020] [Indexed: 12/14/2022]
Abstract
Appropriate human trophoblast lineage specification and differentiation is crucial for the establishment of normal placentation and maintenance of pregnancy. However, due to the lack of proper modeling systems, the molecular mechanisms of these processes are still largely unknown. Much of the early studies in this area have been based on animal models and tumor-derived trophoblast cell lines, both of which are suboptimal for modeling this unique human organ. Recent advances in regenerative and stem cell biology methods have led to development of novel in vitro model systems for studying human trophoblast. These include derivation of human embryonic and induced pluripotent stem cells and establishment of methods for the differentiation of these cells into trophoblast, as well as the more recent derivation of human trophoblast stem cells. In addition, advances in culture conditions, from traditional two-dimensional monolayer culture to 3D culturing systems, have led to development of trophoblast organoid and placenta-on-a-chip model, enabling us to study human trophoblast function in context of more physiologically accurate environment. In this review, we will discuss these various model systems, with a focus on human trophoblast, and their ability to help elucidate the key mechanisms underlying placental development and function. This review focuses on model systems of human trophoblast differentiation, including advantages and limitations of stem cell-based culture, trophoblast organoid, and organ-on-a-chip methods and their applications in understanding placental development and disease.
Collapse
Affiliation(s)
- Mariko Horii
- Department of Pathology, University of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California, USA
| | - Ojeni Touma
- Department of Pathology, University of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California, USA
| | - Tony Bui
- Department of Pathology, University of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
79
|
Baptista LC, Costa ML, Surita FG, Rocha CDS, Lopes-Cendes I, Souza BBD, Costa FF, Melo MBD. Placental transcriptome profile of women with sickle cell disease reveals differentially expressed genes involved in migration, trophoblast differentiation and inflammation. Blood Cells Mol Dis 2020; 84:102458. [PMID: 32562953 DOI: 10.1016/j.bcmd.2020.102458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 12/27/2022]
Abstract
Sickle cell disease (SCD) is a group of disorders whose common characteristic is the presence of hemoglobin (Hb) S in erythrocytes. The main consequence of this abnormality is vaso-occlusion, which can affect almost all organs including the placenta. This study aimed to evaluate the gene expression profile in placentas of women with SCD by means of total RNA sequencing. For this, we proposed a case-control study, with three groups of pregnant women: HbSS (n = 10), HbSC (n = 14) and HbAA (n = 21). The results showed differences in expression in a number of genes such as NOS2 (fold change, FC = 4.52), HLAG (FC = 5.56), ASCL2 (FC = 3.61), CXCL10 (FC = -3.66) and IL1R2 (FC = 3.92) for the HbSC group and S100A8 (FC = -3.82), CPXM2 (FC = 4.57), CXCL10 (FC = -4.59), CXCL11 (FC = -3.72) and CAMP (FC = -4.55) for the HbSS group. Differentially expressed genes are mainly associated with migration, trophoblast differentiation and inflammation. The causes leading to altered gene expression in placentas of sickle cell patients are not fully understood, but the presence of intravascular hemolysis and vaso-occlusion, with cycles of ischemia and reperfusion, may contribute to the emergence of an environment which can be very harmful for placental physiology, altering the nutrient supply and metabolic exchange for fetal growth.
Collapse
Affiliation(s)
- Letícia Carvalho Baptista
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas - UNICAMP, Campinas, SP 13083-875, Brazil.
| | - Maria Laura Costa
- Department of Obstetrics and Gynecology, University of Campinas - UNICAMP, Campinas, SP 13083-880, Brazil.
| | - Fernanda Garanhani Surita
- Department of Obstetrics and Gynecology, University of Campinas - UNICAMP, Campinas, SP 13083-880, Brazil.
| | - Cristiane de Souza Rocha
- Department of Medical Genetics and Genomic Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, SP 13083-887, Brazil.
| | - Iscia Lopes-Cendes
- Department of Medical Genetics and Genomic Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, SP 13083-887, Brazil.
| | - Bruno Batista de Souza
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas - UNICAMP, Campinas, SP 13083-875, Brazil.
| | - Fernando Ferreira Costa
- Hematology and Hemotherapy Center, University of Campinas - UNICAMP, Campinas, SP 13083-878, Brazil.
| | - Mônica Barbosa de Melo
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas - UNICAMP, Campinas, SP 13083-875, Brazil.
| |
Collapse
|
80
|
Nteeba J, Varberg KM, Scott RL, Simon ME, Iqbal K, Soares MJ. Poorly controlled diabetes mellitus alters placental structure, efficiency, and plasticity. BMJ Open Diabetes Res Care 2020; 8:8/1/e001243. [PMID: 32595139 PMCID: PMC7322553 DOI: 10.1136/bmjdrc-2020-001243] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/11/2020] [Accepted: 05/21/2020] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION The hemochorial placenta provides a critical barrier at the maternal-fetal interface to modulate maternal immune tolerance and enable gas and nutrient exchange between mother and conceptus. Pregnancy outcomes are adversely affected by diabetes mellitus; however, the effects of poorly controlled diabetes on placental formation, and subsequently fetal development, are not fully understood. RESEARCH DESIGN AND METHODS Streptozotocin was used to induce hyperglycemia in pregnant rats for the purpose of investigating the impact of poorly controlled diabetes on placental formation and fetal development. The experimental paradigm of hypoxia exposure in the pregnant rat was also used to assess properties of placental plasticity. Euglycemic and hyperglycemic rats were exposed to ambient conditions (~21% oxygen) or hypoxia (10.5% oxygen) beginning on gestation day (gd) 6.5 and sacrificed on gd 13.5. To determine whether the interaction of hyperglycemia and hypoxia was directly altering trophoblast lineage development, rat trophoblast stem (TS) cells were cultured in high glucose (25 mM) and/or exposed to low oxygen (0.5% to 1.5%). RESULTS Diabetes caused placentomegaly and placental malformation, decreasing placental efficiency and fetal size. Elevated glucose disrupted rat TS cell differentiation in vitro. Evidence of altered trophoblast differentiation was also observed in vivo, as hyperglycemia affected the junctional zone transcriptome and interfered with intrauterine trophoblast invasion and uterine spiral artery remodeling. When exposed to hypoxia, hyperglycemic rats showed decreased proliferation and ectoplacental cone development on gd 9.5 and complete pregnancy loss by gd 13.5. Furthermore, elevated glucose concentrations inhibited TS cell responses to hypoxia in vitro. CONCLUSIONS Overall, these results indicate that alterations in placental development, efficiency, and plasticity could contribute to the suboptimal fetal outcomes in offspring from pregnancies complicated by poorly controlled diabetes.
Collapse
Affiliation(s)
- Jackson Nteeba
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Kaela M Varberg
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Regan L Scott
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mikaela E Simon
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Khursheed Iqbal
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Michael J Soares
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, Kansas, USA
- Center for Perinatal Research, Children's Mercy Research Institute, Children's Mercy, Kansas City, Missouri, USA
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
81
|
The Role of NFκB in Healthy and Preeclamptic Placenta: Trophoblasts in the Spotlight. Int J Mol Sci 2020; 21:ijms21051775. [PMID: 32150832 PMCID: PMC7084575 DOI: 10.3390/ijms21051775] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 02/01/2023] Open
Abstract
The NFκB protein family regulates numerous pathways within the cell-including inflammation, hypoxia, angiogenesis and oxidative stress-all of which are implicated in placental development. The placenta is a critical organ that develops during pregnancy that primarily functions to supply and transport the nutrients required for fetal growth and development. Abnormal placental development can be observed in numerous disorders during pregnancy, including fetal growth restriction, miscarriage, and preeclampsia (PE). NFκB is highly expressed in the placentas of women with PE, however its contributions to the syndrome are not fully understood. In this review we discuss the molecular actions and related pathways of NFκB in the placenta and highlight areas of research that need attention.
Collapse
|
82
|
Farah O, Nguyen C, Tekkatte C, Parast MM. Trophoblast lineage-specific differentiation and associated alterations in preeclampsia and fetal growth restriction. Placenta 2020; 102:4-9. [PMID: 33218578 DOI: 10.1016/j.placenta.2020.02.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/06/2020] [Accepted: 02/09/2020] [Indexed: 12/26/2022]
Abstract
The human placenta is a poorly-understood organ, but one that is critical for proper development and growth of the fetus in-utero. The epithelial cell type that contributes to primary placental functions is called "trophoblast," including two main subtypes, villous and extravillous trophoblast. Cytotrophoblast and syncytiotrophoblast comprise the villous compartment and contribute to gas and nutrient exchange, while extravillous trophoblast invade and remodel the uterine wall and vessels, in order to supply maternal blood to the growing fetus. Abnormal differentiation of trophoblast contributes to placental dysfunction and is associated with complications of pregnancy, including preeclampsia (PE) and fetal growth restriction (FGR). This review describes what is known about the cellular organization of the placenta during both normal development and in the setting of PE/FGR. It also explains known trophoblast lineage-specific markers and pathways regulating their differentiation, and how these are altered in the setting of PE/FGR, focusing on studies which have used human placental tissues. Finally, it also highlights remaining questions and needed resources to advance this field.
Collapse
Affiliation(s)
- Omar Farah
- Department of Pathology, University of California San Diego, La Jolla, CA, 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Calvin Nguyen
- Department of Pathology, University of California San Diego, La Jolla, CA, 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Chandana Tekkatte
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, CA, 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
83
|
Treissman J, Yuan V, Baltayeva J, Le HT, Castellana B, Robinson WP, Beristain AG. Low oxygen enhances trophoblast column growth by potentiating differentiation of the extravillous lineage and promoting LOX activity. Development 2020; 147:dev.181263. [PMID: 31871275 DOI: 10.1242/dev.181263] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022]
Abstract
Early placental development and the establishment of the invasive trophoblast lineage take place within a low oxygen environment. However, conflicting and inconsistent findings have obscured the role of oxygen in regulating invasive trophoblast differentiation. In this study, the effect of hypoxic, normoxic and atmospheric oxygen on invasive extravillous pathway progression was examined using a human placental explant model. Here, we show that exposure to low oxygen enhances extravillous column outgrowth and promotes the expression of genes that align with extravillous trophoblast (EVT) lineage commitment. By contrast, supra-physiological atmospheric levels of oxygen promote trophoblast proliferation while simultaneously stalling EVT progression. Low oxygen-induced EVT differentiation coincided with elevated transcriptomic levels of lysyl oxidase (LOX) in trophoblast anchoring columns, in which functional experiments established a role for LOX activity in promoting EVT column outgrowth. The findings of this work support a role for low oxygen in potentiating the differentiation of trophoblasts along the extravillous pathway. In addition, these findings generate insight into new molecular processes controlled by oxygen during early placental development.
Collapse
Affiliation(s)
- Jenna Treissman
- The British Columbia Children's Hospital Research Institute, Vancouver V5Z 4H4, Canada.,Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver V5Z 4H4, Canada
| | - Victor Yuan
- The British Columbia Children's Hospital Research Institute, Vancouver V5Z 4H4, Canada.,Department of Medical Genetics, The University of British Columbia, Vancouver V5Z 4H4, Canada
| | - Jennet Baltayeva
- The British Columbia Children's Hospital Research Institute, Vancouver V5Z 4H4, Canada.,Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver V5Z 4H4, Canada
| | - Hoa T Le
- The British Columbia Children's Hospital Research Institute, Vancouver V5Z 4H4, Canada.,Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver V5Z 4H4, Canada
| | - Barbara Castellana
- The British Columbia Children's Hospital Research Institute, Vancouver V5Z 4H4, Canada.,Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver V5Z 4H4, Canada
| | - Wendy P Robinson
- The British Columbia Children's Hospital Research Institute, Vancouver V5Z 4H4, Canada.,Department of Medical Genetics, The University of British Columbia, Vancouver V5Z 4H4, Canada
| | - Alexander G Beristain
- The British Columbia Children's Hospital Research Institute, Vancouver V5Z 4H4, Canada .,Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver V5Z 4H4, Canada
| |
Collapse
|
84
|
Abstract
The placenta is essential for normal in utero development in mammals. In humans, defective placental formation underpins common pregnancy disorders such as pre-eclampsia and fetal growth restriction. The great variation in placental types across mammals means that animal models have been of limited use in understanding human placental development. However, new tools for studying human placental development, including 3D organoids, stem cell culture systems and single cell RNA sequencing, have brought new insights into this field. Here, we review the morphological, molecular and functional aspects of human placental formation, with a focus on the defining cell of the placenta - the trophoblast.
Collapse
Affiliation(s)
- Margherita Y Turco
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
- Department of Physiology, Neuroscience and Development, University of Cambridge, Cambridge CB2 3EG, UK
| | - Ashley Moffett
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| |
Collapse
|
85
|
Effect of Chlorpyrifos on human extravillous-like trophoblast cells. Reprod Toxicol 2019; 90:118-125. [PMID: 31509763 DOI: 10.1016/j.reprotox.2019.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 09/03/2019] [Accepted: 09/06/2019] [Indexed: 01/11/2023]
Abstract
An increased risk of pregnancy disorders has been reported in women and animal models exposed to organophosphate pesticides. However, less information is available on impacts to human placental function. Here, we addressed the impact of chlorpyrifos (CPF) on extravillous cytotrophoblasts (evCTB) employing HTR8/SVneo cells as an in vitro model. Cell proliferation, migration and invasion were not affected by CPF under conditions where cell viability was not compromised; however, we observed reduced expression of genes for vascular endothelial growth factor receptor 1, hypoxia-inducible factor 1-alpha, peroxisome proliferator activated receptor gamma, and the β-subunit of human chorionic gonadotropin. These results are the first effects reported by organophosphate pesticide in evCTB cells and show altered expression of several genes important for placental development that could serve as potential biomarkers for future research.
Collapse
|
86
|
Expression of glucose transporters in the human amnion derived mesenchymal stromal cells under normoglycemic and hyperglycemic conditions. Biologia (Bratisl) 2019. [DOI: 10.2478/s11756-019-00350-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
87
|
Knöfler M, Haider S, Saleh L, Pollheimer J, Gamage TKJB, James J. Human placenta and trophoblast development: key molecular mechanisms and model systems. Cell Mol Life Sci 2019; 76:3479-3496. [PMID: 31049600 PMCID: PMC6697717 DOI: 10.1007/s00018-019-03104-6] [Citation(s) in RCA: 455] [Impact Index Per Article: 75.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/16/2022]
Abstract
Abnormal placentation is considered as an underlying cause of various pregnancy complications such as miscarriage, preeclampsia and intrauterine growth restriction, the latter increasing the risk for the development of severe disorders in later life such as cardiovascular disease and type 2 diabetes. Despite their importance, the molecular mechanisms governing human placental formation and trophoblast cell lineage specification and differentiation have been poorly unravelled, mostly due to the lack of appropriate cellular model systems. However, over the past few years major progress has been made by establishing self-renewing human trophoblast stem cells and 3-dimensional organoids from human blastocysts and early placental tissues opening the path for detailed molecular investigations. Herein, we summarize the present knowledge about human placental development, its stem cells, progenitors and differentiated cell types in the trophoblast epithelium and the villous core. Anatomy of the early placenta, current model systems, and critical key regulatory factors and signalling cascades governing placentation will be elucidated. In this context, we will discuss the role of the developmental pathways Wingless and Notch, controlling trophoblast stemness/differentiation and formation of invasive trophoblast progenitors, respectively.
Collapse
Affiliation(s)
- Martin Knöfler
- Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, Währinger Gürtel 18-20, 5Q, 1090, Vienna, Austria.
| | - Sandra Haider
- Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, Währinger Gürtel 18-20, 5Q, 1090, Vienna, Austria
| | - Leila Saleh
- Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, Währinger Gürtel 18-20, 5Q, 1090, Vienna, Austria
| | - Jürgen Pollheimer
- Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, Währinger Gürtel 18-20, 5Q, 1090, Vienna, Austria
| | - Teena K J B Gamage
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Joanna James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
88
|
Soares MJ, Varberg KM, Iqbal K. Hemochorial placentation: development, function, and adaptations. Biol Reprod 2019; 99:196-211. [PMID: 29481584 DOI: 10.1093/biolre/ioy049] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/21/2018] [Indexed: 11/12/2022] Open
Abstract
Placentation is a reproductive adaptation that permits fetal growth and development within the protected confines of the female reproductive tract. Through this important role, the placenta also determines postnatal health and susceptibility to disease. The hemochorial placenta is a prominent feature in primate and rodent development. This manuscript provides an overview of the basics of hemochorial placental development and function, provides perspectives on major discoveries that have shaped placental research, and thoughts on strategies for future investigation.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas, USA and the Center for Perinatal Research, Children΄s Research Institute, Children΄s Mercy, Kansas City, Missouri, USA
| | - Kaela M Varberg
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
89
|
Farrell A, Alahari S, Ermini L, Tagliaferro A, Litvack M, Post M, Caniggia I. Faulty oxygen sensing disrupts angiomotin function in trophoblast cell migration and predisposes to preeclampsia. JCI Insight 2019; 4:127009. [PMID: 30996134 DOI: 10.1172/jci.insight.127009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/14/2019] [Indexed: 12/17/2022] Open
Abstract
Human placenta development and a successful pregnancy is incumbent upon precise oxygen-dependent control of trophoblast migration/invasion. Persistent low oxygen leading to failed trophoblast invasion promotes inadequate spiral artery remodeling, a characteristic of preeclampsia. Angiomotin (AMOT) is a multifaceted scaffolding protein involved in cell polarity and migration, yet its upstream regulation and significance in the human placenta remain unknown. Herein, we show that AMOT is primarily expressed in migratory extravillous trophoblast cells (EVTs) of the intermediate and distal anchoring column. Its expression increases after 10 weeks of gestation when oxygen tension rises and EVT migration/invasion peaks. Time-lapse imaging confirmed that the AMOT 80-kDa isoform promotes migration of trophoblastic JEG3 and HTR-8/SVneo cells. In preeclampsia, however, AMOT expression is decreased and its localization to migratory fetomaternal interface EVTs is disrupted. We demonstrate that Jumonji C domain-containing protein 6 (JMJD6), an oxygen sensor, positively regulates AMOT via oxygen-dependent lysyl hydroxylation. Furthermore, in vitro and ex vivo studies show that transforming growth factor-β (TGF-β) regulates AMOT expression, its interaction with polarity protein PAR6, and its subcellular redistribution from tight junctions to cytoskeleton. Our data reveal an oxygen- and TGF-β-driven migratory function for AMOT in the human placenta, and implicate its deficiency in impaired trophoblast migration that plagues preeclampsia.
Collapse
Affiliation(s)
- Abby Farrell
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Institute of Medical Sciences, and
| | - Sruthi Alahari
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Leonardo Ermini
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Andrea Tagliaferro
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Michael Litvack
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Martin Post
- Institute of Medical Sciences, and.,Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Isabella Caniggia
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Institute of Medical Sciences, and.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada.,Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
90
|
Trophoblast-Specific Expression of Hif-1α Results in Preeclampsia-Like Symptoms and Fetal Growth Restriction. Sci Rep 2019; 9:2742. [PMID: 30808910 PMCID: PMC6391498 DOI: 10.1038/s41598-019-39426-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 12/17/2018] [Indexed: 01/02/2023] Open
Abstract
The placenta is an essential organ that is formed during pregnancy and its proper development is critical for embryonic survival. While several animal models have been shown to exhibit some of the pathological effects present in human preeclampsia, these models often do not represent the physiological aspects that have been identified. Hypoxia-inducible factor 1 alpha (Hif-1α) is a necessary component of the cellular oxygen-sensing machinery and has been implicated as a major regulator of trophoblast differentiation. Elevated levels of Hif-1α in the human placenta have been linked to the development of pregnancy-associated disorders, such as preeclampsia and fetal growth restriction. As oxygen regulation is a critical determinant for placentogenesis, we determined the effects of constitutively active Hif-1α, specifically in trophoblasts, on mouse placental development in vivo. Our research indicates that prolonged expression of trophoblast-specific Hif-1α leads to a significant decrease in fetal birth weight. In addition, we noted significant physiological alterations in placental differentiation that included reduced branching morphogenesis, alterations in maternal and fetal blood spaces, and failure to remodel the maternal spiral arteries. These placental alterations resulted in subsequent maternal hypertension with parturitional resolution and maternal kidney glomeruloendotheliosis with accompanying proteinuria, classic hallmarks of preeclampsia. Our findings identify Hif-1α as a critical molecular mediator of placental development and indicate that prolonged expression of Hif-1α, explicitly in placental trophoblasts causes maternal pathology and establishes a mouse model that significantly recapitulates the physiological and pathophysiological characteristics of preeclampsia with fetal growth restriction.
Collapse
|
91
|
Abstract
Preeclampsia, a leading cause of maternal and perinatal morbidity and mortality worldwide, is accompanied by shallow placentation and deficient remodeling of the uterine spiral arteries by invasive placental trophoblast cells during the first trimester of pregnancy. Here, we generated induced pluripotent stem cells from umbilical cords of normal pregnancies and ones complicated by early onset preeclampsia (EOPE) and converted them to trophoblast to recapitulate events of early pregnancy. Parameters disturbed in EOPE, including trophoblast invasiveness, were assessed. Under low O2, both sets of cells behaved similarly, but, under the more stressful 20% O2 conditions, the invasiveness of EOPE trophoblast was markedly reduced. Gene expression changes in EOPE trophoblast suggested a dysregulation invasion linked to high O2. We describe a model for early onset preeclampsia (EOPE) that uses induced pluripotent stem cells (iPSCs) generated from umbilical cords of EOPE and control (CTL) pregnancies. These iPSCs were then converted to placental trophoblast (TB) representative of early pregnancy. Marker gene analysis indicated that both sets of cells differentiated at comparable rates. The cells were tested for parameters disturbed in EOPE, including invasive potential. Under 5% O2, CTL TB and EOPE TB lines did not differ, but, under hyperoxia (20% O2), invasiveness of EOPE TB was reduced. RNA sequencing analysis disclosed no consistent differences in expression of individual genes between EOPE TB and CTL TB under 20% O2, but, a weighted correlation network analysis revealed two gene modules (CTL4 and CTL9) that, in CTL TB, were significantly linked to extent of TB invasion. CTL9, which was positively correlated with 20% O2 (P = 0.02) and negatively correlated with invasion (P = 0.03), was enriched for gene ontology terms relating to cell adhesion and migration, angiogenesis, preeclampsia, and stress. Two EOPE TB modules, EOPE1 and EOPE2, also correlated positively and negatively, respectively, with 20% O2 conditions, but only weakly with invasion; they largely contained the same sets of genes present in modules CTL4 and CTL9. Our experiments suggest that, in EOPE, the initial step precipitating disease is a reduced capacity of placental TB to invade caused by a dysregulation of O2 response mechanisms and that EOPE is a syndrome, in which unbalanced expression of various combinations of genes affecting TB invasion provoke disease onset.
Collapse
|
92
|
Pollheimer J, Vondra S, Baltayeva J, Beristain AG, Knöfler M. Regulation of Placental Extravillous Trophoblasts by the Maternal Uterine Environment. Front Immunol 2018; 9:2597. [PMID: 30483261 PMCID: PMC6243063 DOI: 10.3389/fimmu.2018.02597] [Citation(s) in RCA: 254] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 10/22/2018] [Indexed: 12/22/2022] Open
Abstract
During placentation invasive extravillous trophoblasts (EVTs) migrate into the maternal uterus and modify its vessels. In particular, remodeling of the spiral arteries by EVTs is critical for adapting blood flow and nutrient transport to the developing fetus. Failures in this process have been noticed in different pregnancy complications such as preeclampsia, intrauterine growth restriction, stillbirth, or recurrent abortion. Upon invasion into the decidua, the endometrium of pregnancy, EVTs encounter different maternal cell types such as decidual macrophages, uterine NK (uNK) cells and stromal cells expressing a plethora of growth factors and cytokines. Here, we will summarize development of the EVT lineage, a process occurring independently of the uterine environment, and formation of its different subtypes. Further, we will discuss interactions of EVTs with arteries, veins and lymphatics and illustrate how the decidua and its different immune cells regulate EVT differentiation, invasion and survival. The present literature suggests that the decidual environment and its soluble factors critically modulate EVT function and reproductive success.
Collapse
Affiliation(s)
- Jürgen Pollheimer
- Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| | - Sigrid Vondra
- Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| | - Jennet Baltayeva
- British Columbia's Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
| | - Alexander Guillermo Beristain
- British Columbia's Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
| | - Martin Knöfler
- Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
93
|
Liu H, Kang M, Wang J, Blenkiron C, Lee A, Wise M, Chamley L, Chen Q. Estimation of the burden of human placental micro- and nano-vesicles extruded into the maternal blood from 8 to 12 weeks of gestation. Placenta 2018; 72-73:41-47. [PMID: 30501880 DOI: 10.1016/j.placenta.2018.10.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/18/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND The human placenta extrudes a variety of extracellular vesicles (EVs) into the maternal blood daily. These vesicles may be crucial to the adaptation of the maternal cardiovascular and immune systems to pregnancy. Quantifying the EVs that are released in early gestation is important to our understanding of how placental EVs may contribute to the regulation of maternal physiology. METHODS EVs were isolated from first trimester placental explants and separated into micro- and nano-vesicles by differential centrifugation. The numbers of each type of EVs extruded from each milligram of placentae between gestational weeks 8 and 12 was determined by Nanoparticle Tracking Analysis. The total protein or DNA content of the vesicles was determined by BCA assay or Qubit® 2.0. RESULTS Neither the number of micro- nor nano-EVs/mg explant (n = 49), nor the total protein (n = 19) and DNA content (n = 29) of these EVs changed significantly between 8 and 12 weeks of gestation. When the increasing placental weight with gestation was accounted for, the daily number of placental EVs extruded into the maternal blood increased by more than 100 fold between 8 and 12 weeks (micro-EVs 6.23 X 1014 and nano-EVs 1.84 X 1014 at 12 weeks, p = 0.0003). DISCUSSION Constant production of micro- and nano-EVs per-milligram placenta, regardless of gestational age, and the increased daily burden of EVs across gestational age indicate these EVs have the potential to regulate maternal physiology from early pregnancy. Since total EV protein content, like EV numbers was, constant, this is a potentially reliable surrogate for quantifying EVs.
Collapse
Affiliation(s)
- Haiyan Liu
- The Hospital of Obstetrics & Gynaecology, Fudan University, China; Department of Obstetrics & Gynaecology, The University of Auckland, New Zealand
| | - Matt Kang
- Department of Obstetrics & Gynaecology, The University of Auckland, New Zealand
| | - Julie Wang
- Department of Obstetrics & Gynaecology, The University of Auckland, New Zealand
| | - Cherie Blenkiron
- Department of Obstetrics & Gynaecology, The University of Auckland, New Zealand; Department of Molecular Medicine and Pathology, The University of Auckland, New Zealand
| | - Arier Lee
- Section of Epidemiology and Biostatistics, School of Population Health, University of Auckland, New Zealand
| | - Michelle Wise
- Department of Obstetrics & Gynaecology, The University of Auckland, New Zealand
| | - Larry Chamley
- Department of Obstetrics & Gynaecology, The University of Auckland, New Zealand
| | - Qi Chen
- The Hospital of Obstetrics & Gynaecology, Fudan University, China; Department of Obstetrics & Gynaecology, The University of Auckland, New Zealand.
| |
Collapse
|
94
|
Yang Y, Abdulhasan M, Awonuga A, Bolnick A, Puscheck EE, Rappolee DA. Hypoxic Stress Forces Adaptive and Maladaptive Placental Stress Responses in Early Pregnancy. Birth Defects Res 2018; 109:1330-1344. [PMID: 29105384 DOI: 10.1002/bdr2.1149] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 10/07/2017] [Indexed: 12/19/2022]
Abstract
This review focuses on hypoxic stress and its effects on the placental lineage and the earliest differentiation events in mouse and human placental trophoblast stem cells (TSCs). Although the placenta is a decidual organ at the end of pregnancy, its earliest rapid growth and function at the start of pregnancy precedes and supports growth and function of the embryo. Earliest function requires that TSCs differentiate, however, "hypoxia" supports rapid growth, but not differentiation of TSCs. Most of the literature on earliest placental "hypoxia" studies used 2% oxygen which is normoxic for TSCs. Hypoxic stress happens when oxygen level drops below 2%. It decreases anabolism, proliferation, potency/stemness and increases differentiation, despite culture conditions that would sustain proliferation and potency. Thus, to study the pathogenesis due to TSC dysfunction, it is important to study hypoxic stress below 2%. Many studies have been performed using 0.5 to 1% oxygen in cultured mouse TSCs. From all these studies, a small number has examined human trophoblast lines and primary first trimester placental hypoxic stress responses in culture. Some other stress stimuli, aside from hypoxic stress, are used to elucidate common and unique aspects of hypoxic stress. The key outcomes produced by hypoxic stress are mitochondrial, anabolic, and proliferation arrest, and this is coupled with stemness loss and differentiation. Hypoxic stress can lead to depletion of stem cells and miscarriage, or can lead to later dysfunctions in placentation and fetal development. Birth Defects Research 109:1330-1344, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yu Yang
- CS Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Mohammed Abdulhasan
- CS Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan
| | - Awoniyi Awonuga
- CS Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan
| | - Alan Bolnick
- CS Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan
| | - Elizabeth E Puscheck
- CS Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan
| | - Daniel A Rappolee
- CS Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan.,Institutes for Environmental Health Science, Wayne state University School of Medicine, Detroit, Michigan.,Department of Biology, University of Windsor, Windsor, ON, Canada
| |
Collapse
|
95
|
Soares MJ, Iqbal K, Kozai K. Hypoxia and Placental Development. Birth Defects Res 2018; 109:1309-1329. [PMID: 29105383 DOI: 10.1002/bdr2.1135] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/04/2017] [Indexed: 12/17/2022]
Abstract
Hemochorial placentation is orchestrated through highly regulated temporal and spatial decisions governing the fate of trophoblast stem/progenitor cells. Trophoblast cell acquisition of specializations facilitating invasion and uterine spiral artery remodeling is a labile process, sensitive to the environment, and represents a process that is vulnerable to dysmorphogenesis in pathologic states. Hypoxia is a signal guiding placental development, and molecular mechanisms directing cellular adaptations to low oxygen tension are integral to trophoblast cell differentiation and placentation. Hypoxia can also be used as an experimental tool to investigate regulatory processes controlling hemochorial placentation. These developmental processes are conserved in mouse, rat, and human placentation. Consequently, elements of these developmental events can be modeled and hypotheses tested in trophoblast stem cells and in genetically manipulated rodents. Hypoxia is also a consequence of a failed placenta, yielding pathologies that can adversely affect maternal adjustments to pregnancy, fetal health, and susceptibility to adult disease. The capacity of the placenta for adaptation to environmental challenges highlights the importance of its plasticity in safeguarding a healthy pregnancy. Birth Defects Research 109:1309-1329, 2017.© 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas.,Fetal Health Research, Children's Research Institute, Children's Mercy, Kansas City, Missouri
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Keisuke Kozai
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
96
|
Pham J, Arul Nambi Rajan K, Li P, Parast MM. The role of Sirtuin1-PPARγ axis in placental development and function. J Mol Endocrinol 2018; 60:R201-R212. [PMID: 29467141 PMCID: PMC8584848 DOI: 10.1530/jme-17-0315] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 02/21/2018] [Indexed: 12/11/2022]
Abstract
Placental development is important for proper in utero growth and development of the fetus, as well as maternal well-being during pregnancy. Abnormal differentiation of placental epithelial cells, called trophoblast, is at the root of multiple pregnancy complications, including miscarriage, the maternal hypertensive disorder preeclampsia and intrauterine growth restriction. The ligand-activated nuclear receptor, PPARγ, and nutrient sensor, Sirtuin-1, both play a role in numerous pathways important to cell survival and differentiation, metabolism and inflammation. However, each has also been identified as a key player in trophoblast differentiation and placental development. This review details these studies, and also describes how various stressors, including hypoxia and inflammation, alter the expression or activity of PPARγ and Sirtuin-1, thereby contributing to placenta-based pregnancy complications.
Collapse
Affiliation(s)
- Jonathan Pham
- Department of PathologyUniversity of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative MedicineUniversity of California San Diego, La Jolla, California, USA
| | - Kanaga Arul Nambi Rajan
- Department of PathologyUniversity of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative MedicineUniversity of California San Diego, La Jolla, California, USA
| | - Ping Li
- Department of PathologyMedical School of Jinan University, Guangzhou, China
| | - Mana M Parast
- Department of PathologyUniversity of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative MedicineUniversity of California San Diego, La Jolla, California, USA
| |
Collapse
|
97
|
|
98
|
Hayder H, O'Brien J, Nadeem U, Peng C. MicroRNAs: crucial regulators of placental development. Reproduction 2018; 155:R259-R271. [PMID: 29615475 DOI: 10.1530/rep-17-0603] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 04/03/2018] [Indexed: 12/25/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding single-stranded RNAs that are integral to a wide range of cellular processes mainly through the regulation of translation and mRNA stability of their target genes. The placenta is a transient organ that exists throughout gestation in mammals, facilitating nutrient and gas exchange and waste removal between the mother and the fetus. miRNAs are expressed in the placenta, and many studies have shown that miRNAs play an important role in regulating trophoblast differentiation, migration, invasion, proliferation, apoptosis, vasculogenesis/angiogenesis and cellular metabolism. In this review, we provide a brief overview of canonical and non-canonical pathways of miRNA biogenesis and mechanisms of miRNA actions. We highlight the current knowledge of the role of miRNAs in placental development. Finally, we point out several limitations of the current research and suggest future directions.
Collapse
Affiliation(s)
- Heyam Hayder
- Department of BiologyYork University, Toronto, Ontario, Canada
| | - Jacob O'Brien
- Department of BiologyYork University, Toronto, Ontario, Canada
| | - Uzma Nadeem
- Department of BiologyYork University, Toronto, Ontario, Canada
| | - Chun Peng
- Department of BiologyYork University, Toronto, Ontario, Canada
| |
Collapse
|
99
|
Soncin F, Khater M, To C, Pizzo D, Farah O, Wakeland A, Arul Nambi Rajan K, Nelson KK, Chang CW, Moretto-Zita M, Natale DR, Laurent LC, Parast MM. Comparative analysis of mouse and human placentae across gestation reveals species-specific regulators of placental development. Development 2018; 145:dev.156273. [PMID: 29361559 DOI: 10.1242/dev.156273] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 01/09/2018] [Indexed: 01/08/2023]
Abstract
An increasing body of evidence points to significant spatio-temporal differences in early placental development between mouse and human, but a detailed comparison of placentae in these two species is missing. We set out to compare placentae from both species across gestation, with a focus on trophoblast progenitor markers. We found that CDX2 and ELF5, but not EOMES, are expressed in early post-implantation trophoblast subpopulations in both species. Genome-wide expression profiling of mouse and human placentae revealed clusters of genes with distinct co-expression patterns across gestation. Overall, there was a closer fit between patterns observed in the placentae when the inter-species comparison was restricted to human placentae through gestational week 16 (thus, excluding full-term samples), suggesting that the developmental timeline in mouse runs parallel to the first half of human placental development. In addition, we identified VGLL1 as a human-specific marker of proliferative cytotrophoblast, where it is co-expressed with the transcription factor TEAD4. As TEAD4 is involved in trophectoderm specification in the mouse, we posit a regulatory role for VGLL1 in early events during human placental development.
Collapse
Affiliation(s)
- Francesca Soncin
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| | - Marwa Khater
- Department of Reproductive Medicine, University of California San Diego, La Jolla, CA 92093, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| | - Cuong To
- Department of Reproductive Medicine, University of California San Diego, La Jolla, CA 92093, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| | - Donald Pizzo
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
| | - Omar Farah
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| | - Anna Wakeland
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| | - Kanaga Arul Nambi Rajan
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| | - Katharine K Nelson
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| | - Ching-Wen Chang
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| | - Matteo Moretto-Zita
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| | - David R Natale
- Department of Reproductive Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Louise C Laurent
- Department of Reproductive Medicine, University of California San Diego, La Jolla, CA 92093, USA .,Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA .,Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| |
Collapse
|
100
|
Chen X, Tong C, Li H, Peng W, Li R, Luo X, Ge H, Ran Y, Li Q, Liu Y, Xiong X, Bai Y, Zhang H, Baker PN, Liu X, Qi H. Dysregulated Expression of RPS4Y1 (Ribosomal Protein S4, Y-Linked 1) Impairs STAT3 (Signal Transducer and Activator of Transcription 3) Signaling to Suppress Trophoblast Cell Migration and Invasion in Preeclampsia. Hypertension 2018; 71:481-490. [PMID: 29378854 DOI: 10.1161/hypertensionaha.117.10250] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 09/13/2017] [Accepted: 12/27/2017] [Indexed: 12/15/2022]
Abstract
Normal placentation and a successful pregnancy depend on appropriate trophoblast cell migration and invasion. Inadequate trophoblast invasion and impaired spiral artery remodeling may lead to pregnancy-related disorders, such as preeclampsia. RPS4Y1 (ribosomal protein S4, Y-linked 1) is a member of the S4E family of ribosomal proteins. In this study, we found that RPS4Y1 levels were upregulated in placental samples collected from preeclamptic patients, when compared with the normotensive pregnant women. In vitro, inhibition of RPS4Y1 induced trophoblast cell invasion, promoted placental explant outgrowth, and increased STAT3 (signal transducer and activator of transcription 3) phosphorylation along with elevated expression of N-cadherin and vimentin. Conversely, overexpression of RPS4Y1 results in reduced trophoblast cell invasion and decreased STAT3 phosphorylation. In addition, the suppression of RPS4Y1 promotes trophoblast cell invasion, which could be abolished by the STAT3 knockdown. Meanwhile, we observed reductions of STAT3 phosphorylation expression in preeclampsia patients. Collectively, these results demonstrate that the level of RPS4Y1 expression may be associated with preeclampsia by affecting trophoblast cell migration and invasion via the STAT3/epithelial-mesenchymal transition pathway.
Collapse
Affiliation(s)
- Xuehai Chen
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Chao Tong
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Haiying Li
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Wei Peng
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Rong Li
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Xin Luo
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Huisheng Ge
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Yuxin Ran
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Qin Li
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Yamin Liu
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Xi Xiong
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Yuxiang Bai
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Hua Zhang
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Philip N Baker
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Xiru Liu
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.).
| | - Hongbo Qi
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| |
Collapse
|