Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
Placenta ; 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38879406

ABSTRACT

Correct placental development and function are essential for adapting the mother to the ongoing pregnancy and the wellbeing of the growing fetus; however, underlying processes are still poorly understood. Only limited structural and cellular placental features are shared among species hence requiring reliable human in-vitro models. Recently established trophoblast stem cell and organoid models significantly improved placental research; however, the human placenta constitutes a multi-cellular organ with tightly orchestrated, cellular and molecular networks between trophoblasts (TBs) and villous core cells (VCCs) vital for correct placentation. The establishment of co-culture models is accordingly the logical consequence to investigate TB and VCC interactions, but first requires efficient purification of ideally donor-matched placental cell types. We herein present a meticulously-tailored protocol based on four sequential digestion steps (d-steps) with varying enzyme compositions and digestion mode and length, gently releasing cells layer-by-layer from human first trimester placentae (8 - 9th week of gestation). Using immunofluorescence and flow cytometry, we analyzed the tissue fragments and digestion solutions after every d-step and collected data on individual digestion progress as well as cell viability, counts, and specifications. D-step 1 revealed a significantly low viability and was mainly composed of syncytial fragments, extravillous trophoblasts EVTs, and maternal leukocytes. D-step 2 and 3, comprising high viability predominantly contained TBs (90-99 %) with a significant enrichment of EVTs in d-step 2 and an almost pure villous cytotrophoblast (vCTB) population in d-step 3. D-step 4 finally enabled isolating fetal VCCs consisting of endothelial cells, fibroblasts, and Hofbauer cells. Interestingly, maternal leukocytes were detected in d-step 1 and 2 but completely absent from d-step 3 and 4 revealing pure fetal cell populations. In sum, we present a detailed guideline for stepwise isolating selected placental cell types suitable for further studies and co-culture models investigating TB and VCC interactions involved in early placental development.

2.
J Integr Bioinform ; 20(4)2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38127662

ABSTRACT

During early pregnancy, extravillous trophoblasts (EVTs) play a crucial role in modifying the maternal uterine environment. Failures in EVT lineage formation and differentiation can lead to pregnancy complications such as preeclampsia, fetal growth restriction, and pregnancy loss. Despite recent advances, our knowledge on molecular and external factors that control and affect EVT development remains incomplete. Using trophoblast organoid in vitro models, we recently discovered that coordinated manipulation of the transforming growth factor beta (TGFß) signaling is essential for EVT development. To further investigate gene networks involved in EVT function and development, we performed weighted gene co-expression network analysis (WGCNA) on our RNA-Seq data. We identified 10 modules with a median module membership of over 0.8 and sizes ranging from 1005 (M1) to 72 (M27) network genes associated with TGFß activation status or in vitro culturing, the latter being indicative for yet undiscovered factors that shape the EVT phenotypes. Lastly, we hypothesized that certain therapeutic drugs might unintentionally interfere with placentation by affecting EVT-specific gene expression. We used the STRING database to map correlations and the Drug-Gene Interaction database to identify drug targets. Our comprehensive dataset of drug-gene interactions provides insights into potential risks associated with certain drugs in early gestation.


Subject(s)
Gene Regulatory Networks , Placenta , Pregnancy , Humans , Female , Placenta/metabolism , Trophoblasts/metabolism , Placentation/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
3.
Development ; 150(22)2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37905445

ABSTRACT

Failures in growth and differentiation of the early human placenta are associated with severe pregnancy disorders such as pre-eclampsia and fetal growth restriction. However, regulatory mechanisms controlling development of placental epithelial cells, the trophoblasts, remain poorly elucidated. Using trophoblast stem cells (TSCs), trophoblast organoids (TB-ORGs) and primary cytotrophoblasts (CTBs) of early pregnancy, we herein show that autocrine NOTCH3 signalling controls human placental expansion and differentiation. The NOTCH3 receptor was specifically expressed in proliferative CTB progenitors and its active form, the nuclear NOTCH3 intracellular domain (NOTCH3-ICD), interacted with the transcriptional co-activator mastermind-like 1 (MAML1). Doxycycline-inducible expression of dominant-negative MAML1 in TSC lines provoked cell fusion and upregulation of genes specific for multinucleated syncytiotrophoblasts, which are the differentiated hormone-producing cells of the placenta. However, progenitor expansion and markers of trophoblast stemness and proliferation were suppressed. Accordingly, inhibition of NOTCH3 signalling diminished growth of TB-ORGs, whereas overexpression of NOTCH3-ICD in primary CTBs and TSCs showed opposite effects. In conclusion, the data suggest that canonical NOTCH3 signalling plays a key role in human placental development by promoting self-renewal of CTB progenitors.


Subject(s)
Placenta , Trophoblasts , Humans , Pregnancy , Female , Placenta/metabolism , Receptor, Notch3/genetics , Receptor, Notch3/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Stem Cells , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism
4.
Placenta ; 139: 134-137, 2023 08.
Article in English | MEDLINE | ID: mdl-37390517

ABSTRACT

The human placenta comes in direct contact with maternal cells and blood at two interfaces. The syncytiotrophoblast layer is surrounded by maternal blood at the intervillous space, and extravillous trophoblasts breach the vascular endothelial cells layer upon spiral artery remodeling and invasion of decidual veins. However, little knowledge exists about EVT-derived secreted factors, which may serve as predictive markers for obstetrical syndromes or shape the local environment at the maternal-fetal interface. Here, we define secreted EVT-associated genes and describe a method that yields interstitial fluids from patient-matched first-trimester decidua basalis and parietalis tissues.


Subject(s)
Extracellular Fluid , Placentation , Pregnancy , Female , Humans , Pregnancy Trimester, First , Decidua/metabolism , Endothelial Cells , Trophoblasts/metabolism , Proteins/metabolism
6.
Cell Rep ; 42(1): 111977, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36640334

ABSTRACT

During human pregnancy, placenta-derived extravillous trophoblasts (EVTs) invade the decidua and communicate with maternal immune cells. The decidua distinguishes into basalis (decB) and parietalis (decP). The latter remains unaffected by EVT invasion. By defining a specific gating strategy, we report the accumulation of macrophages in decB. We describe a decidua basalis-associated macrophage (decBAM) population with a differential transcriptome and secretome compared with decidua parietalis-associated macrophages (decPAMs). decBAMs are CD11chi and efficient inducers of Tregs, proliferate in situ, and secrete high levels of CXCL1, CXCL5, M-CSF, and IL-10. In contrast, decPAMs exert a dendritic cell-like, motile phenotype characterized by induced expression of HLA class II molecules, enhanced phagocytosis, and the ability to activate T cells. Strikingly, EVT-conditioned media convert decPAMs into a decBAM phenotype. These findings assign distinct macrophage phenotypes to decidual areas depending on placentation and further highlight a critical role for EVTs in the induction of decB-associated macrophage polarization.


Subject(s)
Decidua , Trophoblasts , Pregnancy , Female , Humans , Pregnancy Trimester, First/physiology , Decidua/metabolism , Trophoblasts/metabolism , Phenotype , Macrophages/metabolism
7.
Placenta ; 133: 19-22, 2023 03 03.
Article in English | MEDLINE | ID: mdl-36696785

ABSTRACT

3-dimensional trophoblast organoids (TB-ORG) represent a reliable model for studying extravillous trophoblast (EVT) lineage formation and differentiation. However, restricted access to first trimester placentae requires alternative cell sources for establishing placental organoids. Recently, we demonstrated EVT differentiation in JEG-3-derived organoids. Consequently, we herein tested whether other commonly used trophoblastic cell lines, ACH-3P, HTR-8/SVneo, and SWAN-71 were capable of self-organizing into organoids and subsequent EVT differentiation. Notably, only ACH-3P formed organoids under stemness conditions mimicking TB-ORG architectures, and induction of EVT differentiation provoked formation of HLA-Gpos areas. Hence ACH-3P-ORGs provide another organoid model for studying controlled EVT lineage formation and differentiation.


Subject(s)
Placenta , Trophoblasts , Pregnancy , Female , Humans , Trophoblasts/metabolism , Placenta/metabolism , Cell Line, Tumor , Pregnancy Trimester, First , Cell Differentiation , Organoids
8.
Proc Natl Acad Sci U S A ; 119(28): e2120667119, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35867736

ABSTRACT

Abnormal placentation has been noticed in a variety of pregnancy complications such as miscarriage, early-onset preeclampsia, and fetal growth restriction. Defects in the developmental program of extravillous trophoblasts (EVTs), migrating from placental anchoring villi into the maternal decidua and its vessels, is thought to be an underlying cause. Yet, key regulatory mechanisms controlling commitment and differentiation of the invasive trophoblast lineage remain largely elusive. Herein, comparative gene expression analyses of HLA-G-purified EVTs, isolated from donor-matched placenta, decidua, and trophoblast organoids (TB-ORGs), revealed biological processes and signaling pathways governing EVT development. In particular, bioinformatics analyses and manipulations in different versatile trophoblast cell models unraveled transforming growth factor-ß (TGF-ß) signaling as a crucial pathway driving differentiation of placental EVTs into decidual EVTs, the latter showing enrichment of a secretory gene signature. Removal of Wingless signaling and subsequent activation of the TGF-ß pathway were required for the formation of human leukocyte antigen-G+ (HLA-G+) EVTs in TB-ORGs that resemble in situ EVTs at the level of global gene expression. Accordingly, TGF-ß-treated EVTs secreted enzymes, such as DAO and PAPPA2, which were predominantly expressed by decidual EVTs. Their genes were controlled by EVT-specific induction and genomic binding of the TGF-ß downstream effector SMAD3. In summary, TGF-ß signaling plays a key role in human placental development governing the differentiation program of EVTs.


Subject(s)
Placentation , Transforming Growth Factor beta , Trophoblasts , Female , HLA-G Antigens/metabolism , Humans , Pregnancy , Transforming Growth Factor beta/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism
9.
Cell Mol Life Sci ; 79(6): 292, 2022 May 13.
Article in English | MEDLINE | ID: mdl-35562545

ABSTRACT

Correct development of the human placenta and its differentiated epithelial cells, syncytial trophoblasts (STBs) and extravillous trophoblasts (EVTs), is crucial for a successful pregnancy outcome. STBs develop by cell fusion of mononuclear cytotrophoblasts (CTBs) in placental floating villi, whereas migratory EVTs originate from specialized villi anchoring to the maternal decidua. Defects in trophoblast differentiation have been associated with severe pregnancy disorders such as early-onset preeclampsia and fetal growth restriction. However, the evolutionary pathways underlying normal and adverse placentation are poorly understood. Herein, we discuss Wingless (WNT) and NOTCH signaling, two pathways that play pivotal roles in human placenta and trophoblast development. Whereas WNT is necessary for expansion of trophoblast progenitors and stem cells, NOTCH1 is required for proliferation and survival of EVT precursors. Differentiation of the latter is orchestrated by a switch in NOTCH receptor expression as well as by changes in WNT ligands and their downstream effectors.


Subject(s)
Placenta , Trophoblasts , Cell Differentiation , Female , Humans , Placenta/metabolism , Placentation , Pregnancy , Receptors, Notch/metabolism
10.
Elife ; 102021 09 03.
Article in English | MEDLINE | ID: mdl-34477104

ABSTRACT

Background: Excessive plasma histamine concentrations cause symptoms in mast cell activation syndrome, mastocytosis, or anaphylaxis. Anti-histamines are often insufficiently efficacious. Human diamine oxidase (hDAO) can rapidly degrade histamine and therefore represents a promising new treatment strategy for conditions with pathological histamine concentrations. Methods: Positively charged amino acids of the heparin-binding motif of hDAO were replaced with polar serine or threonine residues. Binding to heparin and heparan sulfate, cellular internalization and clearance in rodents were examined. Results: Recombinant hDAO is rapidly cleared from the circulation in rats and mice. After mutation of the heparin-binding motif, binding to heparin and heparan sulfate was strongly reduced. The double mutant rhDAO-R568S/R571T showed minimal cellular uptake. The short α-distribution half-life of the wildtype protein was eliminated, and the clearance was significantly reduced in rodents. Conclusions: The successful decrease in plasma clearance of rhDAO by mutations of the heparin-binding motif with unchanged histamine-degrading activity represents the first step towards the development of rhDAO as a first-in-class biopharmaceutical to effectively treat diseases characterized by excessive histamine concentrations in plasma and tissues. Funding: Austrian Science Fund (FWF) Hertha Firnberg program grant T1135 (EG); Sigrid Juselius Foundation, Medicinska Understödsförening Liv och Hälsa rft (TAS and SeV).


Subject(s)
Amine Oxidase (Copper-Containing) , Amino Acid Motifs/genetics , Biological Products , Heparin/metabolism , Histamine Antagonists , Amine Oxidase (Copper-Containing)/chemistry , Amine Oxidase (Copper-Containing)/genetics , Amine Oxidase (Copper-Containing)/metabolism , Animals , Biological Products/chemistry , Biological Products/metabolism , Histamine Antagonists/chemistry , Histamine Antagonists/metabolism , Humans , Mice , Mutation/genetics , Protein Binding/genetics , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
11.
Biol Reprod ; 105(1): 244-257, 2021 07 02.
Article in English | MEDLINE | ID: mdl-33982062

ABSTRACT

The obstetrical conditions placenta accreta spectrum (PAS) and placenta previa are a significant source of pregnancy-associated morbidity and mortality, yet the specific molecular and cellular underpinnings of these conditions are not known. In this study, we identified misregulated gene expression patterns in tissues from placenta previa and percreta (the most extreme form of PAS) compared with control cases. By comparing this gene set with existing placental single-cell and bulk RNA-Seq datasets, we show that the upregulated genes predominantly mark extravillous trophoblasts. We performed immunofluorescence on several candidate molecules and found that PRG2 and AQPEP protein levels are upregulated in both the fetal membranes and the placental disk in both conditions. While this increased AQPEP expression remains restricted to trophoblasts, PRG2 is mislocalized and is found throughout the fetal membranes. Using a larger patient cohort with a diverse set of gestationally aged-matched controls, we validated PRG2 as a marker for both previa and PAS and AQPEP as a marker for only previa in the fetal membranes. Our findings suggest that the extraembryonic tissues surrounding the conceptus, including both the fetal membranes and the placental disk, harbor a signature of previa and PAS that is characteristic of EVTs and that may reflect increased trophoblast invasiveness.


Subject(s)
Eosinophil Major Basic Protein/genetics , Extraembryonic Membranes/metabolism , Gene Expression Regulation , Metalloproteases/genetics , Placenta Accreta/metabolism , Placenta Previa/metabolism , Proteoglycans/genetics , Eosinophil Major Basic Protein/metabolism , Female , Humans , Metalloproteases/metabolism , Pregnancy , Proteoglycans/metabolism
12.
Glycobiology ; 31(4): 444-458, 2021 05 03.
Article in English | MEDLINE | ID: mdl-32985651

ABSTRACT

Human diamine oxidase (hDAO) rapidly inactivates histamine by deamination. No pharmacokinetic data are available to better understand its potential as a new therapeutic modality for diseases with excess local and systemic histamine, like anaphylaxis, urticaria or mastocytosis. After intravenous administration of recombinant hDAO to rats and mice, more than 90% of the dose disappeared from the plasma pool within 10 min. Human DAO did not only bind to various endothelial and epithelial cell lines in vitro, but was also unexpectedly internalized and visible in granule-like structures. The uptake of rhDAO into cells was dependent on neither the asialoglycoprotein-receptor (ASGP-R) nor the mannose receptor (MR) recognizing terminal galactose or mannose residues, respectively. Competition experiments with ASGP-R and MR ligands did not block internalization in vitro or rapid clearance in vivo. The lack of involvement of N-glycans was confirmed by testing various glycosylation mutants. High but not low molecular weight heparin strongly reduced the internalization of rhDAO in HepG2 cells and HUVECs. Human DAO was readily internalized by CHO-K1 cells, but not by the glycosaminoglycan- and heparan sulfate-deficient CHO cell lines pgsA-745 and pgsD-677, respectively. A docked heparin hexasaccharide interacted well with the predicted heparin binding site 568RFKRKLPK575. These results strongly imply that rhDAO clearance in vivo and cellular uptake in vitro is independent of N-glycan interactions with the classical clearance receptors ASGP-R and MR, but is mediated by binding to heparan sulfate proteoglycans followed by internalization via an unknown receptor.


Subject(s)
Amine Oxidase (Copper-Containing) , Heparan Sulfate Proteoglycans , Amine Oxidase (Copper-Containing)/metabolism , Animals , CHO Cells , Cricetinae , Glycosaminoglycans , Heparitin Sulfate/metabolism , Humans , Mice , Rats
13.
Hum Reprod ; 35(11): 2467-2477, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32940686

ABSTRACT

STUDY QUESTION: Do high endothelial venules (HEVs) appear in the uterus of healthy and pathological pregnancies? SUMMARY ANSWER: Our study reveals that HEVs are present in the non-pregnant endometrium and decidua parietalis (decP) but decline upon placentation in decidua basalis (decB) and are less abundant in decidual tissues from idiopathic, recurrent pregnancy losses (RPLs). WHAT IS KNOWN ALREADY: RPL is associated with a compromised decidual vascular phenotype. STUDY DESIGN, SIZE, DURATION: Endometrial (n = 29) and first trimester decidual (n = 86, 6-12th week of gestation) tissue samples obtained from endometrial biopsies or elective pregnancy terminations were used to determine the number of HEVs and T cells. In addition, quantification of HEVs and immune cells was performed in a cohort of decidual tissues from RPL (n = 25). PARTICIPANTS/MATERIALS, SETTING, METHODS: Position and frequency of HEVs were determined in non-pregnant endometrial as well as decidual tissue sections using immunofluorescence (IF) staining with antibodies against E-selectin, intercellular adhesion molecule, von Willebrand factor, ephrin receptor B4, CD34 and a carbohydrate epitope specific to HEVs (MECA-79). Immune cell distribution and characterization was determined by antibodies recognizing CD45 and CD3 by IF staining- and flow cytometry-based analyses. Antibodies against c-c motif chemokine ligand 21 (CCL21) and lymphotoxin-beta were used in IF staining and Western blot analyses of decidual tissues. MAIN RESULTS AND THE ROLE OF CHANCE: Functional HEVs are found in high numbers in the secretory endometrium and decP but decline in numbers upon placentation in decB (P ≤ 0.001). Decidua parietalis tissues contain higher levels of the HEV-maintaining factor lymphotoxin beta and decP-associated HEVs also express CCL21 (P ≤ 0.05), a potent T-cell chemoattractant. Moreover, there is a positive correlation between the numbers of decidual HEVs and the abundance of CD3+ cells in decidual tissue sections (P ≤ 0.001). In-depth analysis of a RPL tissue collection revealed a decreased decB (P ≤ 0.01) and decP (P ≤ 0.01) HEV density as well as reduced numbers of T cells in decB (P ≤ 0.05) and decP (P ≤ .001) sections when compared with age-matched healthy control samples. Using receiver-operating characteristics analyses, we found significant predictive values for the ratios of CD3/CD45 (P < 0.001) and HEVs/total vessels (P < 0.001) for the occurrence of RPL. LIMITATIONS, REASONS FOR CAUTION: Analyses were performed in first trimester decidual tissues from elective terminations of pregnancy or non-pregnant endometrium samples from patients diagnosed with non-endometrial pathologies including cervical polyps, ovarian cysts and myomas. First trimester decidual tissues may include pregnancies which potentially would have developed placental disorders later in gestation. In addition, our cohort of non-pregnant endometrium may not reflect the endometrial vascular phenotype of healthy women. Finally, determination of immune cell distributions in the patient cohorts studied may be influenced by the different modes of tissue derivation. Pregnancy terminations were performed by surgical aspiration, endometrial tissues were obtained by biopsies and RPL tissues were collected after spontaneous loss of pregnancy. WIDER IMPLICATIONS OF THE FINDINGS: In this study, we propose an inherent mechanism by which the endometrium and in particular the decidua control T-cell recruitment. By demonstrating reduced HEV densities and numbers of T cells in decB and decP tissues of RPL samples we further support previous findings reporting an altered vascular phenotype in early pregnancy loss. Altogether, the findings provide important information to further decipher the etiologies of unexplained RPL. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by the Austrian Science Fund (P31470 B30 to M.K.) and by the Austrian National Bank (17613ONB to J.P.). There are no competing interests to declare. TRIAL REGISTRATION NUMBER: N/A.


Subject(s)
Decidua , Trophoblasts , Austria , Female , Humans , Pregnancy , Pregnancy Trimester, First , T-Lymphocytes , Venules
14.
FASEB J ; 34(8): 10720-10730, 2020 08.
Article in English | MEDLINE | ID: mdl-32614494

ABSTRACT

Adequate anchoring of the placenta in the uterus through invasion of first trimester cytotrophoblasts (CTB) is required for a successful pregnancy. This process is mediated by matrix metalloproteinases (MMPs) and regulated by the maternal environment. Obesity is known to alter the intrauterine milieu and has been related to impaired invasion. We hypothesized that placental MMP15, a novel membrane-type MMP, is involved in CTB invasion and regulated by maternal obesity in early pregnancy. Thus, in this study MMP15 was immunolocalized to invasive extravillous and interstitial CTB. MMP15 silencing in chorionic villous explants using two different siRNAs reduced trophoblast outgrowth length (-35%, P ≤ .001 and -26%, P < .05) and area (-43%, P ≤ .001 and -36%, P ≤ .01) without altering trophoblast proliferation or apoptosis. Short-term treatment of primary first trimester trophoblasts with IL-6 (10 ng/mL), interleukin 10 (IL-10) (50 ng/mL), and tumor necrosis factor α (TNF-α) (10 ng/mL) did not affect MMP15 protein levels. Likewise, MMP15 mRNA and protein levels were unaltered between human first trimester placentas from control pregnancies vs those complicated with maternal obesity. Overall, our results suggest that the role of MMP15 in placental development and function in early pregnancy is limited to CTB invasion without being affected by short- and long-term inflammation.


Subject(s)
Cell Movement/physiology , Matrix Metalloproteinase 15/metabolism , Obesity, Maternal/metabolism , Pregnancy Trimester, First/metabolism , Trophoblasts/metabolism , Trophoblasts/physiology , Adult , Apoptosis/physiology , Cell Proliferation/physiology , Cells, Cultured , Female , Humans , Interleukin-10/metabolism , Interleukin-6/metabolism , Male , Placenta/metabolism , Placenta/physiology , Pregnancy , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
Proc Natl Acad Sci U S A ; 117(24): 13562-13570, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32482863

ABSTRACT

Various pregnancy complications, such as severe forms of preeclampsia or intrauterine growth restriction, are thought to arise from failures in the differentiation of human placental trophoblasts. Progenitors of the latter either develop into invasive extravillous trophoblasts, remodeling the uterine vasculature, or fuse into multinuclear syncytiotrophoblasts transporting oxygen and nutrients to the growing fetus. However, key regulatory factors controlling trophoblast self-renewal and differentiation have been poorly elucidated. Using primary cells, three-dimensional organoids, and CRISPR-Cas9 genome-edited JEG-3 clones, we herein show that YAP, the transcriptional coactivator of the Hippo signaling pathway, promotes maintenance of cytotrophoblast progenitors by different genomic mechanisms. Genetic or chemical manipulation of YAP in these cellular models revealed that it stimulates proliferation and expression of cell cycle regulators and stemness-associated genes, but inhibits cell fusion and production of syncytiotrophoblast (STB)-specific proteins, such as hCG and GDF15. Genome-wide comparisons of primary villous cytotrophoblasts overexpressing constitutively active YAP-5SA with YAP KO cells and syncytializing trophoblasts revealed common target genes involved in trophoblast stemness and differentiation. ChIP-qPCR unraveled that YAP-5SA overexpression increased binding of YAP-TEAD4 complexes to promoters of proliferation-associated genes such as CCNA and CDK6 Moreover, repressive YAP-TEAD4 complexes containing the histone methyltransferase EZH2 were detected in the genomic regions of the STB-specific CGB5 and CGB7 genes. In summary, YAP plays a pivotal role in the maintenance of the human placental trophoblast epithelium. Besides activating stemness factors, it also directly represses genes promoting trophoblast cell fusion.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Placentation , Transcription Factors/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Differentiation , Cell Proliferation , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Humans , Muscle Proteins/genetics , Muscle Proteins/metabolism , Placenta/metabolism , Pregnancy , Protein Binding , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , TEA Domain Transcription Factors , Transcription Factors/genetics , YAP-Signaling Proteins
16.
Mol Syst Biol ; 16(4): e9247, 2020 04.
Article in English | MEDLINE | ID: mdl-32323921

ABSTRACT

Prostate cancer (PCa) has a broad spectrum of clinical behavior; hence, biomarkers are urgently needed for risk stratification. Here, we aim to find potential biomarkers for risk stratification, by utilizing a gene co-expression network of transcriptomics data in addition to laser-microdissected proteomics from human and murine prostate FFPE samples. We show up-regulation of oxidative phosphorylation (OXPHOS) in PCa on the transcriptomic level and up-regulation of the TCA cycle/OXPHOS on the proteomic level, which is inversely correlated to STAT3 expression. We hereby identify gene expression of pyruvate dehydrogenase kinase 4 (PDK4), a key regulator of the TCA cycle, as a promising independent prognostic marker in PCa. PDK4 predicts disease recurrence independent of diagnostic risk factors such as grading, staging, and PSA level. Therefore, low PDK4 is a promising marker for PCa with dismal prognosis.


Subject(s)
Gene Expression Profiling/methods , Neoplasm Recurrence, Local/genetics , Neoplasms, Experimental/pathology , Prostatic Neoplasms/genetics , Proteomics/methods , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , STAT3 Transcription Factor/genetics , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Humans , Laser Capture Microdissection , Male , Mice , Neoplasm Grading , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Oxidative Phosphorylation , Prognosis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , STAT3 Transcription Factor/metabolism , Systems Biology , Young Adult
17.
J Lipid Res ; 60(11): 1922-1934, 2019 11.
Article in English | MEDLINE | ID: mdl-31530576

ABSTRACT

During pregnancy, extravillous trophoblasts (EVTs) invade the maternal decidua and remodel the local vasculature to establish blood supply for the growing fetus. Compromised EVT function has been linked to aberrant pregnancy associated with maternal and fetal morbidity and mortality. However, metabolic features of this invasive trophoblast subtype are largely unknown. Using primary human trophoblasts isolated from first trimester placental tissues, we show that cellular cholesterol homeostasis is differentially regulated in EVTs compared with villous cytotrophoblasts. Utilizing RNA-sequencing, gene set-enrichment analysis, and functional validation, we provide evidence that EVTs display increased levels of free and esterified cholesterol. Accordingly, EVTs are characterized by increased expression of the HDL-receptor, scavenger receptor class B type I, and reduced expression of the LXR and its target genes. We further reveal that EVTs express elevated levels of hydroxy-delta-5-steroid dehydrogenase 3 beta- and steroid delta-isomerase 1 (HSD3B1) (a rate-limiting enzyme in progesterone synthesis) and are capable of secreting progesterone. Increasing cholesterol export by LXR activation reduced progesterone secretion in an ABCA1-dependent manner. Importantly, HSD3B1 expression was decreased in EVTs of idiopathic recurrent spontaneous abortions, pointing toward compromised progesterone metabolism in EVTs of early miscarriages. Here, we provide insights into the regulation of cholesterol and progesterone metabolism in trophoblastic subtypes and its putative relevance in human miscarriage.


Subject(s)
Abortion, Habitual/metabolism , Cholesterol/metabolism , Progesterone/metabolism , Trophoblasts/metabolism , Computational Biology , Female , Homeostasis , Humans , Pregnancy , Sequence Analysis, RNA
18.
Cell Mol Life Sci ; 76(18): 3479-3496, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31049600

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.


Subject(s)
Placenta/metabolism , Trophoblasts/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cell Differentiation , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Female , Humans , Models, Biological , Placenta/anatomy & histology , Placentation , Pregnancy , Signal Transduction , Trophoblasts/cytology
19.
Front Immunol ; 9: 2597, 2018.
Article in English | MEDLINE | ID: mdl-30483261

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.


Subject(s)
Placenta/immunology , Placenta/physiology , Trophoblasts/immunology , Trophoblasts/physiology , Uterus/immunology , Uterus/physiology , Animals , Cell Differentiation/immunology , Cell Differentiation/physiology , Cell Movement/immunology , Cell Movement/physiology , Endometrium/immunology , Endometrium/physiology , Female , Humans , Lymphatic Vessels/immunology , Lymphatic Vessels/physiology , Pregnancy
20.
PLoS Genet ; 14(10): e1007698, 2018 10.
Article in English | MEDLINE | ID: mdl-30312291

ABSTRACT

Genome amplification and cellular senescence are commonly associated with pathological processes. While physiological roles for polyploidization and senescence have been described in mouse development, controversy exists over their significance in humans. Here, we describe tetraploidization and senescence as phenomena of normal human placenta development. During pregnancy, placental extravillous trophoblasts (EVTs) invade the pregnant endometrium, termed decidua, to establish an adapted microenvironment required for the developing embryo. This process is critically dependent on continuous cell proliferation and differentiation, which is thought to follow the classical model of cell cycle arrest prior to terminal differentiation. Strikingly, flow cytometry and DNAseq revealed that EVT formation is accompanied with a genome-wide polyploidization, independent of mitotic cycles. DNA replication in these cells was analysed by a fluorescent cell-cycle indicator reporter system, cell cycle marker expression and EdU incorporation. Upon invasion into the decidua, EVTs widely lose their replicative potential and enter a senescent state characterized by high senescence-associated (SA) ß-galactosidase activity, induction of a SA secretory phenotype as well as typical metabolic alterations. Furthermore, we show that the shift from endocycle-dependent genome amplification to growth arrest is disturbed in androgenic complete hydatidiform moles (CHM), a hyperplastic pregnancy disorder associated with increased risk of developing choriocarinoma. Senescence is decreased in CHM-EVTs, accompanied by exacerbated endoreduplication and hyperploidy. We propose induction of cellular senescence as a ploidy-limiting mechanism during normal human placentation and unravel a link between excessive polyploidization and reduced senescence in CHM.


Subject(s)
Cellular Senescence/physiology , Placenta/metabolism , Placenta/physiology , Cell Cycle , Cell Cycle Checkpoints , Cell Differentiation , Cell Movement , Cell Proliferation , Endometrium/cytology , Female , Genome/physiology , Humans , Placentation/genetics , Placentation/physiology , Polyploidy , Pregnancy , Pregnancy Trimester, First , Primary Cell Culture , Tetraploidy , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...