Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 174
Filter
1.
Crit Rev Toxicol ; 54(7): 431-464, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39016688

ABSTRACT

During its temporary tenure, the placenta has extensive and specialized functions that are critical for pre- and post-natal development. The consequences of chemical exposure in utero can have profound effects on the structure and function of pregnancy-associated tissues and the life-long health of the birthing person and their offspring. However, the toxicological importance and critical functions of the placenta to embryonic and fetal development and maturation have been understudied. This narrative will review early placental development in humans and highlight some in vitro models currently in use that are or can be applied to better understand placental processes underlying developmental toxicity due to in utero environmental exposures.


Subject(s)
Placenta , Humans , Pregnancy , Female , Placenta/drug effects , Placentation/drug effects , Models, Biological , Fetal Development/drug effects
2.
Nutrients ; 16(11)2024 May 31.
Article in English | MEDLINE | ID: mdl-38892661

ABSTRACT

Folic acid plays an important role in the synthesis, repair, and methylation of deoxyribonucleic acid (DNA). Currently, most studies have focused on the effects of periconceptional folic acid (FA) supplementation on fetal development, and there is still a lack of population-based research exploring the association between FA use during pregnancy and placental development. This study aimed to investigate the impacts of FA supplementation in different pregnancies on placenta-related parameters at delivery. The study included 2708 pregnant women recruited from Ma'anshan City, Anhui Province, China, between May 2013 and September 2014. Information on FA use from one month before conception to delivery was collected. Placental length, width, and thickness were measured. Multivariable logistic regression analysis was used to assess the effects of FA supplementation in different pregnancies on placenta-related parameters. Based on multiple regression analysis, propensity score weighting was adopted to enhance comparability between different FA supplementation groups. Compared with FA non-users, FA supplementation before conception was associated with increased placental width (0.241 cm, 95%CI: 0.052-0.429, p = 0.013) and increased placental surface area (6.398 cm2, 95%CI: 1.407-11.389, p = 0.012), and FA use in early/middle pregnancy was, respectively, related with increased placental thickness (0.061 cm, 95%CI: 0.004-0.117, p = 0.036; 0.066 cm, 95%CI: 0.004-0.129, p = 0.038). FA use before conception could increase placental width and area, and FA use in early/middle pregnancy could increase placental thickness. To confirm the findings, further investigations are needed.


Subject(s)
Dietary Supplements , Folic Acid , Placenta , Humans , Female , Pregnancy , Folic Acid/administration & dosage , Placenta/drug effects , Adult , China , Placentation/drug effects , Young Adult , Delivery, Obstetric/methods
3.
Front Endocrinol (Lausanne) ; 15: 1386309, 2024.
Article in English | MEDLINE | ID: mdl-38846494

ABSTRACT

Introduction: Leptin and its receptors are expressed by the human placenta throughout gestation, yet the role of leptin in early human placental development is not well characterized. Leptin is overexpressed in the placentas from preeclamptic (PE) pregnancies. PE can result from the impaired invasion of fetal placental cells, cytotrophoblasts (CTBs), into the maternal decidua. We hypothesized that elevated leptin levels would impair human CTB invasion. Methods: The effects of leptin on the invasion of human CTBs were evaluated in three cell models, HTR-8/SVneo cells, primary CTBs, and placental villous explants using invasion assays. Further, leptin receptor expression was characterized in all three cell models using RT-PCR. Further phosphokinase assays were performed in HTR-8/SVneo cells to determine signaling pathways involved in CTB invasion in response to differential leptin doses. Results: We found that, prior to 8 weeks gestation, leptin promoted CTB invasion in the explant model. After 11 weeks gestation in explants, primary CTBs and in HTR-8/SVneo cells, leptin promoted invasion at moderate but not at high concentrations. Further, leptin receptor characterization revealed that leptin receptor expression did not vary over gestation, however, STAT, PI3K and MAPK pathways showed different signaling in response to varied leptin doses. Discussion: These data suggest that the excess placental leptin observed in PE may cause impaired CTB invasion as a second-trimester defect. Leptin's differential effect on trophoblast invasion may explain the role of hyperleptinemia in preeclampsia pathogenesis.


Subject(s)
Gestational Age , Leptin , Receptors, Leptin , Trophoblasts , Humans , Trophoblasts/metabolism , Trophoblasts/drug effects , Trophoblasts/pathology , Leptin/metabolism , Leptin/pharmacology , Female , Pregnancy , Receptors, Leptin/metabolism , Receptors, Leptin/genetics , Placenta/metabolism , Placenta/drug effects , Placenta/pathology , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Dose-Response Relationship, Drug , Signal Transduction , Placentation/drug effects , Cell Movement/drug effects
4.
Redox Biol ; 74: 103238, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38870780

ABSTRACT

Oxidative stress (OS) and endoplasmic reticulum stress (ERS) are at the genesis of placental disorders observed in preeclampsia, intrauterine growth restriction, and maternal hypothyroidism. In this regard, cationic manganese porphyrins (MnPs) comprise potent redox-active therapeutics of high antioxidant and anti-inflammatory potential, which have not been evaluated in metabolic gestational diseases yet. This study evaluated the therapeutic potential of two MnPs, [MnTE-2-PyP]5+ (MnP I) and [MnT(5-Br-3-E-Py)P]5+ (MnP II), in the fetal-placental dysfunction of hypothyroid rats. Hypothyroidism was induced by administration of 6-Propyl-2-thiouracil (PTU) and treatment with MnPs I and II 0.1 mg/kg/day started on the 8th day of gestation (DG). The fetal and placental development, and protein and/or mRNA expression of antioxidant mediators (SOD1, CAT, GPx1), hypoxia (HIF1α), oxidative damage (8-OHdG, MDA), ERS (GRP78 and CHOP), immunological (TNFα, IL-6, IL-10, IL-1ß, IL-18, NLRP3, Caspase1, Gasdermin D) and angiogenic (VEGF) were evaluated in the placenta and decidua on the 18th DG using immunohistochemistry and qPCR. ROS and peroxynitrite (PRX) were quantified by fluorometric assay, while enzyme activities of SOD, GST, and catalase were evaluated by colorimetric assay. MnPs I and II increased fetal body mass in hypothyroid rats, and MnP I increased fetal organ mass. MnPs restored the junctional zone morphology in hypothyroid rats and increased placental vascularization. MnPs blocked the increase of OS and ERS mediators caused by hypothyroidism, showing similar levels of expression of HIFα, 8-OHdG, MDA, Gpx1, GRP78, and Chop to the control. Moreover, MnPs I and/or II increased the protein expression of SOD1, Cat, and GPx1 and restored the expression of IL10, Nlrp3, and Caspase1 in the decidua and/or placenta. However, MnPs did not restore the low placental enzyme activity of SOD, CAT, and GST caused by hypothyroidism, while increased the decidual and placental protein expression of TNFα. The results show that treatment with MnPs improves the fetal-placental development and the placental inflammatory state of hypothyroid rats and protects against oxidative stress and reticular stress caused by hypothyroidism at the maternal-fetal interface.


Subject(s)
Hypothyroidism , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Oxidative Stress , Animals , Pregnancy , Female , Rats , Hypothyroidism/drug therapy , Hypothyroidism/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress/drug effects , Inflammasomes/metabolism , Disease Models, Animal , Placenta/metabolism , Placenta/drug effects , Placentation/drug effects , Antioxidants/pharmacology , Endoplasmic Reticulum Stress/drug effects , Fetal Development/drug effects , Manganese , Metalloporphyrins/pharmacology , Endoplasmic Reticulum Chaperone BiP
5.
Environ Res ; 257: 119159, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38754605

ABSTRACT

Triphenyl phosphate (TPhP) is an organophosphate flame retardant that is widely used in many commercial products. The United States Environmental Protection Agency has listed TPhP as a priority compound that requires health risk assessment. We previously found that TPhP could accumulate in the placentae of mice and impair birth outcomes by activating peroxisome proliferator-activated receptor gamma (PPARγ) in the placental trophoblast. However, the underlying mechanism remains unknown. In this study, we used a mouse intrauterine exposure model and found that TPhP induced preeclampsia (PE)-like symptoms, including new on-set gestational hypertension and proteinuria. Immunofluorescence analysis showed that during placentation, PPARγ was mainly expressed in the labyrinth layer and decidua of the placenta. TPhP significantly decreased placental implantation depth and impeded uterine spiral artery remodeling by activating PPARγ. The results of the in vitro experiments confirmed that TPhP inhibited extravillous trophoblast (EVT) cell migration and invasion by activating PPARγ and inhibiting the PI3K-AKT signaling pathway. Overall, our data demonstrated that TPhP could activate PPARγ in EVT cells, inhibit cell migration and invasion, impede placental implantation and uterine spiral artery remodeling, then induce PE-like symptom and impair birth outcomes. Although the exposure doses used in this study was several orders of magnitude higher than human daily intake, our study highlights the placenta as a potential target organ of TPhP worthy of further research.


Subject(s)
Organophosphates , Placentation , Pre-Eclampsia , Animals , Female , Pregnancy , Pre-Eclampsia/chemically induced , Mice , Placentation/drug effects , Organophosphates/toxicity , Flame Retardants/toxicity , Placenta/drug effects , PPAR gamma/metabolism , PPAR gamma/genetics , Trophoblasts/drug effects , Prenatal Exposure Delayed Effects/chemically induced
6.
Toxicology ; 504: 153796, 2024 May.
Article in English | MEDLINE | ID: mdl-38582279

ABSTRACT

As a broad-spectrum and efficient insecticide, beta-Cypermethrin (ß-CYP) poses a health risk to pregnancy. It matters the mechanisms of maternal exposure to ß-CYP for impacting reproductive health. The placenta, a transient organ pivotal for maternal-fetal communication during pregnancy, plays a crucial role in embryonic development. The effect of ß-CYP exposure on the placenta and its underlying molecular mechanisms remain obscure. The objective of this study was to investigate the effect of ß-CYP exposure on placental development and the function of trophoblast, as well as the underlying mechanisms through CD-1 mouse model (1, 10, 20 mg/kg.bw) and in vitro HTR-8/SVneo cell model (12.5, 25, 50, 100 µM). We found slower weight gain and reduced uterine wet weight in pregnant mice with maternal exposure to ß-CYP during pregnancy, as well as adverse pregnancy outcomes such as uterine bleeding and embryo resorption. The abnormal placental development in response to ß-CYP was noticed, including imbalanced placental structure and disrupted labyrinthine vascular development. Trophoblasts, pivotal in placental development and vascular remodeling, displayed abnormal differentiation under ß-CYP exposure. This aberration was characterized by thickened trophoblast layers in the labyrinthine zone, accompanied by mitochondrial and endoplasmic reticulum swelling within trophoblasts. Further researches on human chorionic trophoblast cell lines revealed that ß-CYP exposure induced apoptosis in HTR-8/SVneo cells. This induction resulted in a notable decrease in migration and invasion abilities, coupled with oxidative stress and the inhibition of the Notch signaling pathway. N-acetylcysteine (an antioxidant) partially restored the impaired Notch signaling pathway in HTR-8/SVneo cells, and mitigated cellular functional damage attributed to ß-CYP exposure. Collectively, exposure to ß-CYP induced oxidative stress and then led to inhibition of the Notch signaling pathway and dysfunction of trophoblast cells, ultimately resulted in abnormal placenta and pregnancy. These findings indicate Reactive Oxygen Species as potential intervention targets to mitigate ß-CYP toxicity. The comprehensive elucidation contributes to our understanding of ß-CYP biosafety and offers an experimental basis for preventing and managing its reproductive toxicity.


Subject(s)
Insecticides , Oxidative Stress , Pyrethrins , Trophoblasts , Pyrethrins/toxicity , Female , Pregnancy , Trophoblasts/drug effects , Trophoblasts/pathology , Trophoblasts/metabolism , Oxidative Stress/drug effects , Animals , Mice , Insecticides/toxicity , Humans , Maternal Exposure/adverse effects , Placentation/drug effects , Cell Line , Placenta/drug effects , Placenta/pathology , Placenta/metabolism , Apoptosis/drug effects
7.
Int J Mol Sci ; 22(20)2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34681648

ABSTRACT

Type 1 diabetes mellitus (T1DM) is associated with reduced fetal growth in early pregnancy, but a contributing role of the placenta has remained elusive. Thus, we investigated whether T1DM alters placental development in the first trimester. Using a protein array, the level of 60 cell-cycle-related proteins was determined in human first trimester placental tissue (gestational week 5-11) from control (n = 11) and T1DM pregnancies (n = 12). Primary trophoblasts (gestational week 7-12, n = 32) were incubated in the absence (control) or presence of hyperglycemia (25 mM D-glucose) and hyperosmolarity (5.5 mM D-glucose + 19.5 mM D-mannitol). We quantified the number of viable and dead trophoblasts (CASY Counter) and assessed cell cycle distribution (FACS) and trophoblast invasion using a transwell assay. T1DM was associated with a significant (p < 0.05) downregulation of Ki67 (-26%), chk1 (-25%), and p73 (-26%). The number of viable trophoblasts was reduced under hyperglycemia (-23%) and hyperosmolarity (-18%), whereas trophoblast invasion was increased only under hyperglycemia (+6%). Trophoblast cell death and cell cycle distribution remained unaffected. Collectively, our data demonstrate that hyperglycemia decreases trophoblast proliferation as a potential contributing factor to the reduced placental growth in T1DM in vivo.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Diabetes Mellitus, Type 1/pathology , Glucose/pharmacology , Placenta/metabolism , Adult , Cell Cycle Checkpoints/drug effects , Cell Movement/drug effects , Down-Regulation/drug effects , Female , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Mannitol/pharmacology , Placentation/drug effects , Pregnancy , Pregnancy Trimester, First , Trophoblasts/cytology , Trophoblasts/drug effects , Trophoblasts/metabolism
8.
Endocrinology ; 162(12)2021 12 01.
Article in English | MEDLINE | ID: mdl-34647996

ABSTRACT

Iron deficiency, which occurs when iron demands chronically exceed intake, is prevalent in pregnant women. Iron deficiency during pregnancy poses major risks for the baby, including fetal growth restriction and long-term health complications. The placenta serves as the interface between a pregnant mother and her baby, and it ensures adequate nutrient provisions for the fetus. Thus, maternal iron deficiency may impact fetal growth and development by altering placental function. We used a rat model of diet-induced iron deficiency to investigate changes in placental growth and development. Pregnant Sprague-Dawley rats were fed either a low-iron or iron-replete diet starting 2 weeks before mating. Compared with controls, both maternal and fetal hemoglobin were reduced in dams fed low-iron diets. Iron deficiency decreased fetal liver and body weight, but not brain, heart, or kidney weight. Placental weight was increased in iron deficiency, due primarily to expansion of the placental junctional zone. The stimulatory effect of iron deficiency on junctional zone development was recapitulated in vitro, as exposure of rat trophoblast stem cells to the iron chelator deferoxamine increased differentiation toward junctional zone trophoblast subtypes. Gene expression analysis revealed 464 transcripts changed at least 1.5-fold (P < 0.05) in placentas from iron-deficient dams, including altered expression of genes associated with oxygen transport and lipoprotein metabolism. Expression of genes associated with iron homeostasis was unchanged despite differences in levels of their encoded proteins. Our findings reveal robust changes in placentation during maternal iron deficiency, which could contribute to the increased risk of fetal distress in these pregnancies.


Subject(s)
Iron Deficiencies/physiopathology , Placentation/physiology , Pregnancy Complications/physiopathology , Trophoblasts/physiology , Animals , Cell Differentiation/drug effects , Diet , Dietary Supplements , Female , Iron/pharmacology , Iron/therapeutic use , Iron Deficiencies/complications , Iron Deficiencies/diet therapy , Maternal-Fetal Exchange/drug effects , Placentation/drug effects , Pregnancy , Pregnancy Complications/diet therapy , Rats , Rats, Sprague-Dawley , Trophoblasts/drug effects
9.
Genes (Basel) ; 12(8)2021 08 17.
Article in English | MEDLINE | ID: mdl-34440429

ABSTRACT

Uterine spiral artery remodeling is essential for placental perfusion and fetal growth and, when impaired, results in placental ischemia and pregnancy complications, e.g., fetal growth restriction, preeclampsia, premature birth. Despite the high incidence of adverse pregnancies, current treatment options are limited. Accordingly, research has shifted to the development of gene therapy technologies that provide targeted delivery of "payloads" to the placenta while limiting maternal and fetal exposure. This review describes the current strategies, including placental targeting peptide-bound liposomes, nanoparticle or adenovirus constructs decorated with specific peptide sequences and placental gene promoters delivered via maternal IV injection, directly into the placenta or the uterine artery, as well as noninvasive site-selective targeting of regulating genes conjugated with microbubbles via contrast-enhanced ultrasound. The review also provides a perspective on the effectiveness of these technologies in various animal models and their practicability and potential use for targeted placental delivery of therapeutics and genes in adverse human pregnancies affected by placental dysfunction.


Subject(s)
Fetal Growth Retardation/therapy , Genetic Therapy , Peptides/genetics , Placentation/genetics , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/pathology , Humans , Liposomes/therapeutic use , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Peptides/therapeutic use , Placenta/drug effects , Placenta/physiology , Placentation/drug effects , Pregnancy , Uterus/drug effects , Uterus/growth & development
10.
Sci Rep ; 11(1): 16602, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34400704

ABSTRACT

Di(2-ethylhexyl) phthalate (DEHP) is a synthetic chemical commonly used for its plasticizing capabilities. Because of the extensive production and use of DEHP, humans are exposed to this chemical daily. Diet is a significant exposure pathway and fatty food contain the highest level of phthalates. The impact on pregnancy following DEHP exposure and the associated interaction of high fat (HF) diet remains unknown. Here we report that exposure of pregnant mice to an environmentally relevant level of DEHP did not affect pregnancy. In contrast, mice fed a HF diet during gestation and exposed to the same level of DEHP display marked impairment in placental development, resulting in poor pregnancy outcomes. Our study further reveals that DEHP exposure combined with a HF diet interfere with the signaling pathway controlled by nuclear receptor PPARγ to adversely affect differentiation of trophoblast cells, leading to compromised vascularization and glucose transport in the placenta. Collectively, these findings demonstrate that maternal diet during pregnancy is a critical factor that determines whether exposure to an environmental toxicant results in impaired placental and fetal development, causing intrauterine growth restriction, fetal morbidity, and mortality.


Subject(s)
Diet, High-Fat/adverse effects , Diethylhexyl Phthalate/toxicity , Environmental Pollutants/toxicity , Placentation/drug effects , Animals , Cell Differentiation/drug effects , Estrogens/blood , Female , Fetal Growth Retardation/etiology , Gestational Age , Glucose/metabolism , Glucose Transporter Type 1/metabolism , Mice , PPAR gamma/physiology , Placenta/metabolism , Pregnancy , Pregnancy Outcome , Progesterone/blood , Signal Transduction/drug effects , Trophoblasts/cytology , Trophoblasts/drug effects , Trophoblasts/metabolism
11.
Am J Physiol Cell Physiol ; 321(4): C694-C703, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34406903

ABSTRACT

The human placenta is of vital importance for proper nutrient and waste exchange, immune regulation, and overall fetal health and growth. Specifically, the extracellular matrix (ECM) of placental syncytiotrophoblasts, which extends outward from the placental chorionic villi into maternal blood, acts on a molecular level to regulate and maintain this barrier. Importantly, placental barrier dysfunction has been linked to diseases of pregnancy such as preeclampsia and intrauterine growth restriction. To help facilitate our understanding of the interface and develop therapeutics to repair or prevent dysfunction of the placental barrier, in vitro models of the placental ECM would be of great value. In this study, we aimed to characterize the ECM of an in vitro model of the placental barrier using syncytialized BeWo choriocarcinoma cells. Syncytialization caused a marked change in syndecans, integral proteoglycans of the ECM, which matched observations of in vivo placental ECM. Syndecan-1 expression increased greatly and predominated the other variants. Barrier function of the ECM, as measured by electric cell-substrate impedance sensing (ECIS), increased significantly during and after syncytialization, whereas the ability of THP-1 monocytes to adhere to syncytialized BeWos was greatly reduced compared with nonsyncytialized controls. Furthermore, ECIS measurements indicated that ECM degradation with matrix metalloproteinase-9 (MMP-9), but not heparanase, decreased barrier function. This decrease in ECIS-measured barrier function was not associated with any changes in THP-1 adherence to syncytialized BeWos treated with heparanase or MMP-9. Thus, syncytialization of BeWos provides a physiologically accurate placental ECM with a barrier function matching that seen in vivo.


Subject(s)
Extracellular Matrix/metabolism , Placentation , Syndecan-1/metabolism , Trophoblasts/metabolism , Cell Movement , Electric Impedance , Extracellular Matrix/drug effects , Female , Glucuronidase/pharmacology , Humans , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/pharmacology , Monocytes/metabolism , Permeability , Placentation/drug effects , Pregnancy , Syndecan-1/genetics , THP-1 Cells , Trophoblasts/drug effects , Up-Regulation
12.
Front Endocrinol (Lausanne) ; 12: 694885, 2021.
Article in English | MEDLINE | ID: mdl-34394001

ABSTRACT

Background: Per- and polyfluoroalkyl substances (PFAS) are persistent organic pollutants that have become globally ubiquitous in humans and the environment. In utero PFAS exposure is associated with neurodevelopmental effects; however, the mechanism is poorly understood. Brain-derived neurotrophic factor (BDNF) signaling is critical to fetal neurodevelopment during pregnancy and maintains important regulatory roles later in life. This study aims to characterize placental BDNF signaling and investigate whether PFAS exposure disrupts the signaling pathway in placental trophoblast cells. Methods: The expression and localization of BDNF receptors-p75NTR and TrkB-in first trimester and term human placentas and trophoblast cells were investigated by immunofluorescence staining. To assess the effects of PFAS exposure on the BDNF pathway, BeWo cells were treated with PFAS mixtures that mimicked blood levels in a highly exposed population and major PFAS compounds in the mixture at 0.01, 0.1, 1, and 10 µM concentrations. Changes in pro-BDNF levels and phosphorylation of TrkB receptors were examined by Western blot. Results: In first trimester human placentas, TrkB and p75NTR receptors were primarily localized to syncytiotrophoblast and cytotrophoblast cells. At term, TrkB and p75NTR receptors were primarily observed in the placental villous stroma. TrkB receptor staining in trophoblasts was reduced at term, while p75NTR receptor staining was negative. TrkB receptors were confined to the nuclear and perinuclear spaces, and phosphorylation occurred at the Tyr816 residue in BeWo cells. Exposure to PFOS, PFOA, PFBS, and the six-PFAS mixture did not significantly affect BDNF levels or activation (phosphorylation) of TrkB. Treating cells with 1 µM and 10 µM of PFNA resulted in increased TrkB phosphorylation compared to unexposed controls, but BDNF levels were unchanged. Conclusions: BDNF receptors are present in different regions of human placental villi, indicating diverse functions of BDNF signaling in placental development. Our findings suggest that the BDNF pathway in placental trophoblast cells is not disrupted by exposures to PFOS, PFOA, PFBS, and a PFAS mixture, but may be affected by PFNA exposures. Further investigation is needed on how PFAS affects other critical signaling pathways during fetal neurodevelopment.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Fluorocarbons/toxicity , Trophoblasts/drug effects , Female , Humans , Maternal Exposure/adverse effects , Membrane Glycoproteins/metabolism , Phosphorylation , Placenta/drug effects , Placenta/metabolism , Placentation/drug effects , Pregnancy , Pregnancy Trimester, First/drug effects , Pregnancy Trimester, First/metabolism , Receptor, trkB/metabolism , Signal Transduction/drug effects , Trophoblasts/metabolism , Tumor Cells, Cultured
13.
Mol Nutr Food Res ; 65(18): e2100301, 2021 09.
Article in English | MEDLINE | ID: mdl-34289236

ABSTRACT

INTRODUCTION: As an exogenous food contaminant, dietary oxidized lipid impairs growth and development, and triggers chronic diseases in humans or animals. This study explores the effects of soybean oil with different oxidative degree on the placental injury of gestational rats. METHODS AND RESULTS: Thirty-two female adult rats are randomly assigned to four groups. The control group is fed the purified diet with fresh soybean oil (FSO), and the treatment groups are fed purified diets with lipid content replaced by oxidized soybean oil (OSO) at 200, 400, and 800 mEqO2 kg-1 from conception until delivery. On day 20 of gestation, OSO decreased placental and embryonic weights as the oxidative degree increased linearly and quadratically. The expression of Bax showed a linear increase, and Bcl-2 decreased as the oxidative degree increased. The expression of Fosl1 and Esx1 is linearly and quadratically decreased in OSO-treated groups than FSO group. OSO decreased the level of IL-10 but increased expression of IL-1ß in placenta and plasma. OSO remarkably upregulates levels of Fatp1 and Glut1 and decreases expression of Snat2 and Glut3. CONCLUSION: OSO aggravates placental injury by modulating nutrient transporters and apoptosis-related genes, impedes placental growth and development, and ultimately leads to the decrease of fetal weight.


Subject(s)
Carrier Proteins/metabolism , Maternal Exposure/adverse effects , Placenta/drug effects , Soybean Oil/adverse effects , Soybean Oil/chemistry , Amino Acid Transport System A/metabolism , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Cytokines/blood , Cytokines/genetics , Fatty Acid Transport Proteins/metabolism , Fatty Acid-Binding Proteins/metabolism , Female , Fetal Weight/drug effects , Gene Expression Regulation, Developmental/drug effects , Glucose Transporter Type 3/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Oxidative Stress/physiology , Placenta/metabolism , Placenta/pathology , Placentation/drug effects , Pregnancy , Rats, Sprague-Dawley
14.
Diabetes ; 70(10): 2250-2263, 2021 10.
Article in English | MEDLINE | ID: mdl-34315726

ABSTRACT

Choline metabolite trimethylamine N-oxide (TMAO) has been recognized as a risk factor of gestational diabetes mellitus (GDM), but its exact role in GDM has not been reported. In this study, we focused on the placenta development to reveal the role of TMAO in GDM. We found that the TMAO levels in peripheral and cord plasma were increased in women with GDM and that TMAO levels were positively correlated with newborn weight and placental thickness. Neutrophil extracellular traps (NETs) in the peripheral and cord plasma and the myeloperoxidase expression in the placenta of women with GDM also increased. NETs could inhibit the proliferation, migration, invasion, and angiogenesis of HTR-8/Svneo cells. However, TMAO not only could inhibit the formation of NETs but also could enhance the biological function of HTR-8/Svneo cells. With induction of GDM in NETs-deficient PAD4-/- and wild-type mice, the placental weight of PAD4-/- mice increased significantly. TMAO feeding also inhibited the formation of NETs and further increased the weight of the placenta and fetuses, and this increase did not affect the placental structure. Our data indicate that higher TMAO levels and the formation of abnormal NETs were associated with GDM. TMAO not only could promote the development of the placenta and fetuses but also could inhibit the formation of NETs.


Subject(s)
Diabetes, Gestational/physiopathology , Extracellular Traps/drug effects , Methylamines/pharmacology , Placentation/drug effects , Adult , Animals , Case-Control Studies , Cells, Cultured , Choline/metabolism , Diabetes, Gestational/pathology , Extracellular Traps/metabolism , Female , Fetal Development/drug effects , Fetal Development/genetics , Humans , Infant, Newborn , Methylamines/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Protein-Arginine Deiminase Type 4/genetics , Young Adult
15.
Cell Death Dis ; 12(7): 635, 2021 06 21.
Article in English | MEDLINE | ID: mdl-34155192

ABSTRACT

FURIN is a pro-protein convertase previously shown to be important for placental syncytialisation (Zhou et al. [1]), a process of cell fusion whereby placental cytotrophoblast cells fuse to form a multinucleated syncytium. This finding has been broadly accepted however, we have evidence suggesting the contrary. Spontaneously syncytialising term primary human trophoblast cells and BeWo choriocarcinoma cells were treated with either FURIN siRNA or negative control siRNA or the protease inhibitor, DEC-RVKR-CMK, or vehicle. Cells were then left to either spontaneously syncytialise (primary trophoblasts) or were induced to syncytialise with forskolin (BeWo). Effects on syncytialisation were measured by determining human chorionic gonadotrophin secretion and E-cadherin protein levels. We showed that FURIN is not important for syncytialisation in either cell type. However, in primary trophoblasts another protease also inhibited by DEC-RVKR-CMK, may be involved. Our results directly contrast with those published by Zhou et al. Zhou et al. however, used first trimester villous explants to study syncytialisation, and we used term primary trophoblasts. Therefore, we suggest that FURIN may be involved in syncytialisation of first trimester trophoblasts, but not term trophoblasts. What is more concerning is that our results using BeWo cells do not agree with their results, even though for the most part, we used the same experimental design. It is unclear why these experiments yielded different results, however we wanted to draw attention to simple differences in measuring syncytialisation or flaws in method reporting (including omission of cell line source and passage numbers, siRNA concentration and protein molecular weights) and choice of immunoblot loading controls, that could impact on experimental outcomes. Our study shows that careful reporting of methods by authors and thorough scrutiny by referees are vital. Furthermore, a universal benchmark for measuring syncytialisation is required so that various studies of syncytialisation can be validated.


Subject(s)
Cell Fusion , Furin/metabolism , Placentation , Trophoblasts/enzymology , Amino Acid Chloromethyl Ketones/pharmacology , Antigens, CD/metabolism , Cadherins/metabolism , Cell Line, Tumor , Chorionic Gonadotropin/metabolism , Colforsin/pharmacology , Female , Furin/antagonists & inhibitors , Furin/genetics , Humans , Placentation/drug effects , Pregnancy , Pregnancy Trimester, First , Serine Proteinase Inhibitors/pharmacology , Term Birth , Trophoblasts/drug effects
16.
Reprod Toxicol ; 103: 1-17, 2021 08.
Article in English | MEDLINE | ID: mdl-34015474

ABSTRACT

Proper placental development and function relies on hormone receptors and signaling pathways that make the placenta susceptible to disruption by endocrine disrupting chemicals, such as phthalates. Here, we review relevant research on the associations between phthalate exposures and dysfunctions of the development and function of the placenta, including morphology, physiology, and genetic and epigenetic effects. This review covers in vitro studies, in vivo studies in mammals, and studies in humans. We also discuss important gaps in the literature. Overall, the evidence indicates that toxicity to the placental and maternal-fetal interface is associated with exposure to phthalates. Further studies are needed to better elucidate the mechanisms through which phthalates act in the placenta as well as additional human studies that assess placental disruption through pregnancy with larger sample sizes.


Subject(s)
Phthalic Acids/toxicity , Placenta/drug effects , Placentation/drug effects , Animals , Endocrine Disruptors/toxicity , Female , Humans , Maternal Exposure , Pregnancy
17.
Reprod Biomed Online ; 42(3): 481-504, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33549483

ABSTRACT

RESEARCH QUESTION: Maternal alcohol consumption produces fetal retardation and malformations, probably associated with placental defects. Does perigestational alcohol consumption up to organogenesis lead to abnormal placentation and embryo growth restriction by disrupting the vascular endothelial growth factor (VEGF) system in embryo-placental development? DESIGN: Female mice were treated with 10% ethanol in drinking water before and up to day 10 of gestation. Control mice received ethanol-free water. After treatment, the trophoblastic tissue, embryo growth and the angiogenic VEGF pathway were analysed. RESULTS: Female mice who had received treatment had resorbed and delayed implantation sites with poor ectoplacental cone development. Reduced trophoblastic area tissue from female mice who had received treatment had abnormal junctional zone and diminished labyrinthine vascularization. After treatment, the labyrinth had increased chorionic trophoblast proliferation, hypoxia inducible factor-1α immunoexpression but reduced apoptosis. The embryo growth was reduced concomitantly with low VEGF immunostaining but high endothelial nitric oxide synthase (eNOS) expression. In junctional and labyrinth of treated female mice, gene and protein immunoexpression of VEGF was reduced and the protein expression of FLT-1 increased compared with controls. Increased activation of kinase insert domain receptor receptor (phosphorylated KDR) and expression of eNOS were observed in placenta of treated female mice. Immunoexpression of metalloproteinase-9, however, was reduced in junctional zone but increased in labyrinth, compared with controls. CONCLUSIONS: These data reveal inadequate expression of VEGF/receptors and angiogenic eNOS and metalloproteinase factors related to abnormal early placentation after perigestational alcohol ingestion, providing insight into aetiological factors underlying early placentopathy associated with intrauterine growth restriction caused by maternal alcohol consumption.


Subject(s)
Alcohol Drinking/adverse effects , Central Nervous System Depressants/adverse effects , Embryonic Development/drug effects , Ethanol/adverse effects , Placentation/drug effects , Abortion, Spontaneous/chemically induced , Alcohol Drinking/metabolism , Animals , Female , Fetal Growth Retardation/chemically induced , Matrix Metalloproteinases/metabolism , Mice , Neovascularization, Physiologic/drug effects , Nitric Oxide Synthase Type III/metabolism , Pregnancy , Trophoblasts/drug effects , Vascular Endothelial Growth Factor A/metabolism
18.
Sci Rep ; 11(1): 4029, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33597628

ABSTRACT

Prenatal cannabis use is a significant problem and poses important health risks for the developing fetus. The molecular mechanisms underlying these changes are not fully elucidated but are thought to be attributed to delta-9-tetrahydrocannabinol (THC), the main bioactive constituent of cannabis. It has been reported that THC may target the mitochondria in several tissue types, including placental tissue and trophoblast cell lines, and alter their function. In the present study, in response to 48-h THC treatment of the human extravillous trophoblast cell line HTR8/SVneo, we demonstrate that cell proliferation and invasion are significantly reduced. We further demonstrate THC-treatment elevated levels of cellular reactive oxygen species and markers of lipid damage. This was accompanied by evidence of increased mitochondrial fission. We also observed increased expression of cellular stress markers, HSP70 and HSP60, following exposure to THC. These effects were coincident with reduced mitochondrial respiratory function and a decrease in mitochondrial membrane potential. Taken together, our results suggest that THC can induce mitochondrial dysfunction and reduce trophoblast invasion; outcomes that have been previously linked to poor placentation. We also demonstrate that these changes in HTR8/SVneo biology may be variably mediated by cannabinoid receptors CB1 and CB2.


Subject(s)
Dronabinol/adverse effects , Mitochondria/drug effects , Trophoblasts/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Chaperonin 60/drug effects , Chaperonin 60/genetics , Dronabinol/pharmacology , Female , HSP70 Heat-Shock Proteins/drug effects , HSP70 Heat-Shock Proteins/genetics , Humans , Mitochondria/physiology , Mitochondrial Dynamics , Placenta/metabolism , Placentation/drug effects , Pregnancy , Reactive Oxygen Species
19.
Am J Physiol Endocrinol Metab ; 320(3): E527-E538, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33427051

ABSTRACT

Insulin-like growth factor-1 (IGF-1) is an important fetal growth factor. However, the role of fetal IGF-1 in increasing placental blood flow, nutrient transfer, and nutrient availability to support fetal growth and protein accretion is not well understood. Catheterized fetuses from late gestation pregnant sheep received an intravenous infusion of LR3 IGF-1 (LR3 IGF-1; n = 8) or saline (SAL; n = 8) for 1 wk. Sheep then underwent a metabolic study to measure uterine and umbilical blood flow, nutrient uptake rates, and fetal protein kinetic rates. By the end of the infusion, fetal weights were not statistically different between groups (SAL: 3.260 ± 0.211 kg, LR3 IGF-1: 3.682 ± 0.183; P = 0.15). Fetal heart, adrenal gland, and spleen weights were higher (P < 0.05), and insulin was lower in LR3 IGF-1 (P < 0.05). Uterine and umbilical blood flow and umbilical uptake rates of glucose, lactate, and oxygen were similar between groups. Umbilical amino acid uptake rates were lower in LR3 IGF-1 (P < 0.05) as were fetal concentrations of multiple amino acids. Fetal protein kinetic rates were similar. LR3 IGF-1 skeletal muscle had higher myoblast proliferation (P < 0.05). In summary, LR3 IGF-1 infusion for 1 wk into late gestation fetal sheep increased the weight of some fetal organs. However, because umbilical amino acid uptake rates and fetal plasma amino acid concentrations were lower in the LR3 IGF-1 group, we speculate that animals treated with LR3 IGF-1 can efficiently utilize available nutrients to support organ-specific growth in the fetus rather than by stimulating placental blood flow or nutrient transfer to the fetus.NEW & NOTEWORTHY After a 1-wk infusion of LR3 IGF-1, late gestation fetal sheep had lower umbilical uptake rates of amino acids, lower fetal arterial amino acid and insulin concentrations, and lower fetal oxygen content; however, LR-3 IGF-1-treated fetuses were still able to effectively utilize the available nutrients and oxygen to support organ growth and myoblast proliferation.


Subject(s)
Fetal Development/drug effects , Insulin-Like Growth Factor I/pharmacology , Nutrients/metabolism , Animals , Energy Metabolism/drug effects , Female , Fetal Blood/metabolism , Fetal Weight/drug effects , Fetus/drug effects , Fetus/metabolism , Insulin-Like Growth Factor I/administration & dosage , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/embryology , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Organ Size/drug effects , Placenta/drug effects , Placentation/drug effects , Pregnancy , Sheep
20.
Placenta ; 103: 152-155, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33126049

ABSTRACT

Cytotrophoblast cells fuse to form the syncytiotrophoblast, the main structure responsible for the placenta's specialized functions. This complex process denominated syncytialization is fundamental for a correct pregnancy outcome. We observed that the endocannabinoid anandamide disrupts syncytialization employing traditional techniques and flow cytometry in BeWo cell line.


Subject(s)
Endocannabinoids/pharmacology , Trophoblasts/drug effects , Arachidonic Acids/pharmacology , Cell Fusion/methods , Cell Line, Tumor , Colforsin/pharmacology , Female , Flow Cytometry , Humans , Placentation/drug effects , Polyunsaturated Alkamides/pharmacology , Pregnancy , Signal Transduction/drug effects , Trophoblasts/cytology , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL