Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 308
Filter
Add more filters

Publication year range
1.
Development ; 148(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34559199

ABSTRACT

The therian-specific gene paternally expressed 10 (Peg10) plays an essential role in placenta formation: Peg10 knockout mice exhibit early embryonic lethality as a result of severe placental defects. The PEG10 protein exhibits homology with long terminal repeat (LTR) retrotransposon GAG and POL proteins; therefore, we generated mice harboring a mutation in the highly conserved viral aspartic protease motif in the POL-like region of PEG10 because this motif is essential for the life cycle of LTR retrotransposons/retroviruses. Intriguingly, frequent perinatal lethality, not early embryonic lethality, was observed with fetal and placental growth retardation starting mid-gestation. In the mutant placentas, severe defects were observed in the fetal vasculature, where PEG10 is expressed in the three trophoblast cell layers that surround fetal capillary endothelial cells. Thus, Peg10 has essential roles, not only in early placenta formation, but also in placental vasculature maintenance from mid- to late-gestation. This implies that along the feto-maternal placenta interface an interaction occurs between two retrovirus-derived genes, Peg10 and retrotransposon Gag like 1 (Rtl1, also called Peg11), that is essential for the maintenance of fetal capillary endothelial cells.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Capillaries/metabolism , DNA-Binding Proteins/metabolism , Placenta/blood supply , RNA-Binding Proteins/metabolism , Amino Acid Motifs , Animals , Apoptosis Regulatory Proteins/chemistry , Capillaries/embryology , DNA-Binding Proteins/chemistry , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Mice , Placenta/embryology , Pregnancy , Pregnancy Proteins/chemistry , Pregnancy Proteins/metabolism , RNA-Binding Proteins/chemistry
2.
BMC Pregnancy Childbirth ; 23(1): 725, 2023 Oct 11.
Article in English | MEDLINE | ID: mdl-37821857

ABSTRACT

BACKGROUND: The placenta serves as the sole maternal organ responsible for transmitting nutrients to the fetus, playing a crucial role in supporting standard fetal growth and development. To date, only a small number of studies have investigated the impact of maternal gestational weight gain and lipid concentrations on placental development. This study aimed to explore the influence of weight gain during pregnancy and lipid levels in the second trimester on placental weight, volume, and the placental weight ratio. METHODS: This birth cohort study encompassed 1,358 mother-child pairs. Placental data for each participant was gathered immediately post-delivery, and the study incorporated data on gestational weight gain throughout pregnancy and lipid profiles from the mid-trimester. A linear regression model was employed to assess the correlations between gestational weight gain, mid-trimester lipid levels, and metrics such as placental weight, placental volume, and the placental-to-birth weight ratio (PFR). RESULTS: In the study groups of pre-pregnancy underweight, normal weight, and overweight, the placental weight increased by 4.93 g (95% CI: 1.04-8.81), 2.52 g (95% CI: 1.04-3.99), and 3.30 g (95% CI: 0.38-6.22) per 1 kg of gestational weight gain, respectively. Within the pre-pregnancy underweight and normal weight groups, the placental volume increased by 6.79 cm^3 (95% CI: 3.43-10.15) and 2.85 cm^3 (95% CI: 1.31-4.39) per 1 kg of gestational weight gain, respectively. Additionally, placental weight exhibited a positive correlation with triglyceride (TG) levels (ß = 9.81, 95% CI: 3.28-16.34) and a negative correlation with high-density lipoprotein (HDL-C) levels (ß = - 46.30, 95% CI: - 69.49 to - 23.11). Placental volume also showed a positive association with TG levels (ß = 14.54, 95% CI: 7.69-21.39). Conversely, PFR demonstrated a negative correlation with increasing HDL-C levels (ß = - 0.89, 95% CI: - 1.50 to - 0.27). CONCLUSIONS: Gestational weight gain was significantly correlated with both placental weight and volume. This association was especially pronounced in women who, prior to pregnancy, were underweight or of normal weight. Additionally, TG and HDL-C levels during the mid-trimester were linked to placental development.


Subject(s)
Gestational Weight Gain , Placenta , Female , Humans , Pregnancy , Birth Weight , Cohort Studies , East Asian People , Lipids , Placenta/embryology , Thinness , Organ Size
3.
PLoS Genet ; 16(4): e1008739, 2020 04.
Article in English | MEDLINE | ID: mdl-32320395

ABSTRACT

Inositol 1,4,5-trisphosphate receptors (IP3Rs) are a family of intracellular Ca2+ release channels located on the ER membrane, which in mammals consist of 3 different subtypes (IP3R1, IP3R2, and IP3R3) encoded by 3 genes, Itpr1, Itpr2, and Itpr3, respectively. Studies utilizing genetic knockout mouse models have demonstrated that IP3Rs are essential for embryonic survival in a redundant manner. Deletion of both IP3R1 and IP3R2 has been shown to cause cardiovascular defects and embryonic lethality. However, it remains unknown which cell types account for the cardiovascular defects in IP3R1 and IP3R2 double knockout (DKO) mice. In this study, we generated conditional IP3R1 and IP3R2 knockout mouse models with both genes deleted in specific cardiovascular cell lineages. Our results revealed that deletion of IP3R1 and IP3R2 in cardiomyocytes by TnT-Cre, in endothelial / hematopoietic cells by Tie2-Cre and Flk1-Cre, or in early precursors of the cardiovascular lineages by Mesp1-Cre, resulted in no phenotypes. This demonstrated that deletion of both IP3R genes in cardiovascular cell lineages cannot account for the cardiovascular defects and embryonic lethality observed in DKO mice. We then revisited and performed more detailed phenotypic analysis in DKO embryos, and found that DKO embryos developed cardiovascular defects including reduced size of aortas, enlarged cardiac chambers, as well as growth retardation at embryonic day (E) 9.5, but in varied degrees of severity. Interestingly, we also observed allantoic-placental defects including reduced sizes of umbilical vessels and reduced depth of placental labyrinth in DKO embryos, which could occur independently from other phenotypes in DKO embryos even without obvious growth retardation. Furthermore, deletion of both IP3R1 and IP3R2 by the epiblast-specific Meox2-Cre, which targets all the fetal tissues and extraembryonic mesoderm but not extraembryonic trophoblast cells, also resulted in embryonic lethality and similar allantoic-placental defects. Taken together, our results demonstrated that IP3R1 and IP3R2 play an essential and redundant role in maintaining the integrity of fetal-maternal connection and embryonic viability.


Subject(s)
Fetal Growth Retardation/genetics , Fetal Heart/metabolism , Heart Defects, Congenital/genetics , Inositol 1,4,5-Trisphosphate Receptors/genetics , Placenta/metabolism , Animals , Endothelial Progenitor Cells/metabolism , Female , Fetal Heart/embryology , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Placenta/embryology , Pregnancy
4.
Radiology ; 298(2): 403-412, 2021 02.
Article in English | MEDLINE | ID: mdl-33231529

ABSTRACT

Background Prenatal identification of placenta accreta spectrum (PAS) disorder is essential for treatment planning. More objective means for predicting PAS and clinical outcome may be provided by MRI descriptors. Purpose To investigate the association of intraplacental fetal vessel (IFV) diameter at MRI with PAS and peripartum complications. Materials and Methods Between March 2016 and October 2019, 160 gravid women suspected of having PAS underwent placental MRI as part of a prospective trial. Secondary analysis was performed by two experienced genitourinary radiologists for presence and maximum diameter of IFVs. Relative risk ratios were computed to test the association of IFVs with presence and depth of PAS invasiveness. Receiver operating characteristic analysis was used to evaluate the ability of IFV diameter to help predict PAS, placenta percreta, and peripartum complications and for comparison of the area under the curve (AUC) versus that from other combined MRI predictors of PAS (eg, myometrial thinning, intraplacental T2-hypointense bands, uterine bulge, serosal hypervascularity, and signs of extrauterine placental spread). Intraoperative and histopathologic findings were the reference standard. Results A total of 155 women were evaluated (mean age, 35 years ± 5 [standard deviation]; mean gestational age, 32 weeks ± 3). PAS was diagnosed in 126 of 155 women (81%) (placental percreta in 68 of 126 [54%]). At delivery, 30 of 126 women (24%) experienced massive blood loss (>2000 mL). IFVs were detected at MRI in 109 of 126 women with PAS (86%) and in 67 of 68 women with placental percreta (98%). The relative risk ratio was 2.4 (95% CI: 1.6, 3.4; P < .001) for PAS and 10 (95% CI: 1.5, 70.4; P < .001) for placental percreta when IFVs were visible. IFVs of 2 mm or greater were associated with PAS (AUC, 0.81; 95% CI: 0.67, 0.95; P = .04). IVFs of 3 mm or greater were associated with placenta percreta (AUC, 0.81; 95% CI: 0.73, 0.89; P < .001) and with peripartum complications, including massive bleeding (AUC, 0.80; 95% CI: 0.71, 0.89; P < .001). Combining assessment of IFVs with other MRI descriptors improved the ability of MRI to predict PAS (AUC, 0.94 vs 0.89; P = .009). Conclusion Assessment of intraplacental fetal vessels with other MRI descriptors improved the ability of MRI to help predict PAS. Vessel diameter of 3 mm or greater was predictive of placenta percreta and peripartum complications. © RSNA, 2020 Online supplemental material is available for this article. See also the editorial by Dighe in this issue.


Subject(s)
Magnetic Resonance Imaging/methods , Placenta Accreta/diagnosis , Placenta/blood supply , Placenta/embryology , Prenatal Diagnosis/methods , Adult , Female , Humans , Placenta/diagnostic imaging , Pregnancy , Prospective Studies
5.
Genes Cells ; 25(6): 427-438, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32267063

ABSTRACT

All trophoblast subtypes of the placenta are derived from trophoblast stem cells (TSCs). TSCs have the capacity to self-renew, but how the proliferation of these cells is regulated in the undifferentiated state has been largely unclear. We now show that the F-box protein Skp2 regulates the proliferation of TSCs and thereby plays a pivotal role in placental development in mice on the C57BL/6 background. The placenta of Skp2-/- mouse embryos on the C57BL/6 background was smaller than that of their Skp2+/+ littermates, with the mutant embryos also manifesting intrauterine growth retardation. Although the Skp2-/- mice were born alive, most of them died before postnatal day 21, presumably as a result of placental defects. Depletion of Skp2 in TSCs cultured in the undifferentiated state resulted in a reduced rate of proliferation and arrest of the cell cycle in G1 phase, indicative of a defect in self-renewal capacity. The cell cycle arrest apparent in Skp2-deficient TSCs was reversed by additional ablation of the cyclin-dependent kinase inhibitor (CKI) p57 but not by that of the CKI p27. Our results thus suggest that Skp2-mediated degradation of p57 is an important determinant of the self-renewal capacity of TSCs during placental development, at least in mice of certain genetic backgrounds.


Subject(s)
Cell Cycle/genetics , Embryo, Mammalian/metabolism , Placenta/metabolism , Placentation/genetics , S-Phase Kinase-Associated Proteins/metabolism , Stem Cells/metabolism , Trophoblasts/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinase Inhibitor p57/genetics , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Embryo, Mammalian/embryology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Placenta/embryology , Pregnancy , Rats , S-Phase Kinase-Associated Proteins/genetics
6.
Cytokine ; 143: 155517, 2021 07.
Article in English | MEDLINE | ID: mdl-33814270

ABSTRACT

Vertical transmission of Toxoplasma gondii leads to adverse pregnancy outcomes depending on the time at which the infection occurs and the immunological state of the mother. C57BL/6 and BALB/c mice have been described as susceptible and resistant mouse lineages to congenital T. gondii infection, respectively. This study aimed to elucidate the systemic and local cytokine profile of pregnant mice infected with T. gondii and whether the expression of the transcription factor FOXP3, related to T regulatory cells, is associated with the resistance/susceptibility of these lineages of mice in the context of experimental congenital toxoplasmosis. For this purpose, C57BL/6 and BALB/c females were orally infected with the T. gondii ME-49 strain on the day of vaginal plug detection or day 14 of gestation, examined 7 or 5 days later, respectively, as models of early and late pregnancy. Cytokine levels were measured systemically and in the uterus/placenta. Additionally, the uterus/placenta were evaluated macroscopically for resorption rates and histologically for parasite and FOXP3 immunostaining. The FOXP3 protein expression was also evaluated by western blotting assay. It was found that, during early pregnancy, the infection leads to high IFN-γ, TNF and IL-6 levels systemically, with the TNF levels being higher in C57BL/6 mice. At the maternal-fetal interface, the infection induced high levels of IFN-γ in both mouse lineages; however, higher levels were observed in BALB/c, while high TNF and IL-6 levels were found in C57BL/6, but not in BALB/c mice. In contrast, in late gestation, T. gondii interfered less strongly with the cytokine profile. In early pregnancy, a reduction of FOXP3 expression at the maternal-fetal interface of infected mice was also observed, and the reduction was larger in C57BL/6 compared with BALB/c mice. Additionally, the parasite was seldom found in the uterus/placenta. Thus, the worse pregnancy outcomes observed in C57BL/6 mice were associated with higher TNF systemically, and TNF and IL-6 at the maternal-fetal interface, with lower FOXP3 expression.


Subject(s)
Forkhead Transcription Factors/metabolism , Interleukin-6/blood , Maternal-Fetal Exchange , Pregnancy Outcome , Toxoplasmosis, Congenital/blood , Tumor Necrosis Factor-alpha/blood , Animals , Disease Models, Animal , Female , Interferon-gamma/blood , Lung/parasitology , Mice, Inbred BALB C , Mice, Inbred C57BL , Parasites/physiology , Placenta/embryology , Placenta/metabolism , Placenta/parasitology , Pregnancy , Toxoplasma/physiology , Toxoplasmosis, Animal/blood , Uterus/embryology , Uterus/pathology
7.
Development ; 144(13): 2392-2401, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28576770

ABSTRACT

The placental labyrinth is the interface for gas and nutrient exchange between the embryo and the mother; hence its proper development is essential for embryogenesis. However, the molecular mechanism underlying development of the placental labyrinth, particularly in terms of its endothelial organization, is not well understood. Here, we determined that fibronectin leucine-rich transmembrane protein 2 (FLRT2), a repulsive ligand of the UNC5 receptor family for neurons, is unexpectedly expressed in endothelial cells specifically in the placental labyrinth. Mice lacking FLRT2 in endothelial cells exhibited embryonic lethality at mid-gestation, with systemic congestion and hypoxia. Although they lacked apparent deformities in the embryonic vasculature and heart, the placental labyrinths of these embryos exhibited aberrant alignment of endothelial cells, which disturbed the feto-maternal circulation. Interestingly, this vascular deformity was related to endothelial repulsion through binding to the UNC5B receptor. Our results suggest that the proper organization of the placental labyrinth depends on coordinated inter-endothelial repulsion, which prevents uncontrolled layering of the endothelium.


Subject(s)
Membrane Glycoproteins/metabolism , Organogenesis , Placenta/embryology , Placenta/metabolism , Signal Transduction , Animals , Cell Survival , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Endothelial Cells/metabolism , Female , Gene Deletion , Hypoxia/pathology , Membrane Glycoproteins/deficiency , Mice, Inbred C57BL , Neovascularization, Physiologic , Netrin Receptors , Placenta/blood supply , Placenta/cytology , Pregnancy , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/metabolism
8.
Development ; 144(13): 2469-2479, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28526753

ABSTRACT

EGFL7 is a secreted angiogenic factor produced by embryonic endothelial cells. To understand its role in placental development, we established a novel Egfl7 knockout mouse. The mutant mice have gross defects in chorioallantoic branching morphogenesis and placental vascular patterning. Microangiography and 3D imaging revealed patchy perfusion of Egfl7-/- placentas marked by impeded blood conductance through sites of narrowed vessels. Consistent with poor feto-placental perfusion, Egfl7 knockout resulted in reduced placental weight and fetal growth restriction. The placentas also showed abnormal fetal vessel patterning and over 50% reduction in fetal blood space. In vitro, placental endothelial cells were deficient in migration, cord formation and sprouting. Expression of genes involved in branching morphogenesis, Gcm1, Syna and Synb, and in patterning of the extracellular matrix, Mmrn1, were temporally dysregulated in the placentas. Egfl7 knockout did not affect expression of the microRNA embedded within intron 7. Collectively, these data reveal that Egfl7 is crucial for placental vascularization and embryonic growth, and may provide insight into etiological factors underlying placental pathologies associated with intrauterine growth restriction, which is a significant cause of infant morbidity and mortality.


Subject(s)
Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Neovascularization, Physiologic , Perfusion , Placenta/blood supply , Placenta/embryology , Placentation , Proteins/metabolism , Animals , Base Sequence , Blood Proteins/genetics , Blood Proteins/metabolism , Body Patterning , Calcium-Binding Proteins , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Movement , Down-Regulation/genetics , EGF Family of Proteins , Endothelial Cells/metabolism , Female , Fetal Blood/metabolism , Fetus/embryology , Fetus/metabolism , Gene Expression Regulation, Developmental , Mice, Inbred C57BL , Mice, Knockout , Organ Size , Placenta/metabolism , Pregnancy
9.
J Assist Reprod Genet ; 37(12): 3057-3067, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33089439

ABSTRACT

PURPOSE: This study aimed to determine the effects of drilling and thinning treatment of laser-assisted hatching on the expression and methylation of imprinted gene IGF2/H19 in embryos and offspring. METHODS: The prehatching blastocysts with treatment of drilling or thinning, or control prehatching blastocysts, were transplanted in surrogate uteri. The DNA methylation of IGF2/H19 imprinting control region (ICR) and the expression of IGF2 and H19 were respectively evaluated using bisulfite conversion-mediated sequencing and real-time PCR. RESULTS: The drilling group showed a significant increase in the development rate of hatched blastocysts in comparison with the control and thinning group. DNA methylation level of IGF2/H19 ICR of hatched blastocysts in the thinning group was 27.33% in comparison with the 38.67% and 36% observed in the control and drilling group. The thinning treatment increased the DNA methylation level of IGF2/H19 ICR in the placenta in comparison with the control and drilling group. The drilling and thinning treatment decreased the expression level of H19 mRNA in prehatching and hatched blastocysts as well as placenta, while a significant increase in the expression level of H19 mRNA of offspring was observed in the thinning group. The thinning treatment increased the expression level of IGF2 mRNA of prehatching blastocysts and offspring and a significant decrease in placenta, while the drilling treatment resulted in a significant increase in the expression level of IGF2 mRNA of hatched blastocysts and placenta. CONCLUSION: These observations suggested that drilling used for hatching of in vitro cultured mouse blastocysts may improve the production of offspring.


Subject(s)
Blastocyst/physiology , DNA Methylation , Fertilization in Vitro/methods , Genomic Imprinting , Insulin-Like Growth Factor II/metabolism , Placenta/embryology , RNA, Long Noncoding/genetics , Animals , Animals, Newborn , Blastocyst/cytology , Blastocyst/radiation effects , Embryo Culture Techniques , Female , Insulin-Like Growth Factor II/genetics , Lasers , Male , Mice , Pregnancy
10.
Int J Mol Sci ; 21(20)2020 Oct 14.
Article in English | MEDLINE | ID: mdl-33066440

ABSTRACT

Placental homeostasis of tryptophan is essential for fetal development and programming. The two main metabolic pathways (serotonin and kynurenine) produce bioactive metabolites with immunosuppressive, neurotoxic, or neuroprotective properties and their concentrations in the fetoplacental unit must be tightly regulated throughout gestation. Here, we investigated the expression/function of key enzymes/transporters involved in tryptophan pathways during mid-to-late gestation in rat placenta and fetal organs. Quantitative PCR and heatmap analysis revealed the differential expression of several genes involved in serotonin and kynurenine pathways. To identify the flux of substrates through these pathways, Droplet Digital PCR, western blot, and functional analyses were carried out for the rate-limiting enzymes and transporters. Our findings show that placental tryptophan metabolism to serotonin is crucial in mid-gestation, with a subsequent switch to fetal serotonin synthesis. Concurrently, at term, the close interplay between transporters and metabolizing enzymes of both placenta and fetal organs orchestrates serotonin homeostasis and prevents hyper/hypo-serotonemia. On the other hand, the placental production of kynurenine increases during pregnancy, with a low contribution of fetal organs throughout gestation. Any external insult to this tightly regulated harmony of transporters and enzymes within the fetoplacental unit may affect optimal in utero conditions and have a negative impact on fetal programming.


Subject(s)
Fetus/metabolism , Placenta/metabolism , Transcriptome , Tryptophan/metabolism , Animals , Female , Fetus/embryology , Gene Expression Regulation, Developmental , Metabolic Networks and Pathways , Placenta/embryology , Pregnancy , Rats , Rats, Wistar , Tryptophan/genetics
11.
Int J Mol Sci ; 21(10)2020 May 21.
Article in English | MEDLINE | ID: mdl-32455665

ABSTRACT

Placental disorders are a major cause of pregnancy loss in humans, and 40-60% of embryos are lost between fertilization and birth. Successful embryo implantation and placental development requires rapid proliferation, invasion, and migration of trophoblast cells. In recent years, microRNAs (miRNAs) have emerged as key regulators of molecular pathways involved in trophoblast function. A miRNA binds its target mRNA in the 3'-untranslated region (3'-UTR), causing its degradation or translational repression. Lethal-7 (let-7) miRNAs induce cell differentiation and reduce cell proliferation by targeting proliferation-associated genes. The oncoprotein LIN28 represses the biogenesis of mature let-7 miRNAs. Proliferating cells have high LIN28 and low let-7 miRNAs, whereas differentiating cells have low LIN28 and high let-7 miRNAs. In placenta, low LIN28 and high let-7 miRNAs can lead to reduced proliferation of trophoblast cells, resulting in abnormal placental development. In trophoblast cells, let-7 miRNAs reduce the expression of proliferation factors either directly by binding their mRNA in 3'-UTR or indirectly by targeting the AT-rich interaction domain (ARID)3B complex, a transcription-activating complex comprised of ARID3A, ARID3B, and histone demethylase 4C (KDM4C). In this review, we discuss regulation of trophoblast function by miRNAs, focusing on the role of LIN28-let-7-ARID3B pathway in placental development.


Subject(s)
DNA-Binding Proteins/genetics , MicroRNAs/genetics , Placenta/metabolism , RNA-Binding Proteins/genetics , Animals , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Developmental , Humans , MicroRNAs/metabolism , Placenta/embryology , Pregnancy , RNA-Binding Proteins/metabolism
12.
Scand J Immunol ; 90(5): e12756, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30739345

ABSTRACT

Pre-eclampsia (PE) is a leading cause of maternal and perinatal morbidity and mortality; however, the aetiology of PE still remains unclear. It has been widely accepted that the disease results from insufficient spiral artery remodelling, leading to placental ischaemia and the release of a variety of factors. In recent decades, a large number of studies have observed an abnormal immune response in preeclamptic women and studies of both patients and animal models have shown alterations in the function or the number of immune agents. Thus, researchers believe that alterations in the immune system may contribute to the genesis and pathophysiology of PE. Therefore, identifying the role of the immune system can not only shed light on the nature of PE but also contribute to the development of diagnostic and therapeutic methods for PE. This review focuses on the current knowledge of the immune system including both innate and adaptive immunity and sheds light on their role in PE. Additionally, advances in potential therapeutic measures are discussed.


Subject(s)
Adaptive Immunity/immunology , Immunity, Innate/immunology , Placenta/pathology , Pre-Eclampsia/immunology , Pre-Eclampsia/pathology , B-Lymphocytes, Regulatory/immunology , Female , Humans , Immune Tolerance/immunology , Placenta/embryology , Pregnancy , Th1-Th2 Balance , Trophoblasts/cytology
13.
Am J Obstet Gynecol ; 221(3): 273.e1-273.e9, 2019 09.
Article in English | MEDLINE | ID: mdl-31226291

ABSTRACT

BACKGROUND: Fetal growth restriction accounts for a significant proportion of perinatal morbidity and death. The cerebroplacental ratio is gaining much interest as a useful tool in differentiating the "at-risk" fetus in both fetal growth restriction and appropriate-for-gestational-age pregnancies. The Prospective Observational Trial to Optimize Pediatric Health in Fetal Growth Restriction group has demonstrated previously that the presence of this "brain-sparing" effect is associated significantly with adverse perinatal outcomes in the fetal growth restriction cohort. However, data about neurodevelopment in children from pregnancies that are complicated by fetal growth restriction are sparse and conflicting. OBJECTIVE: The aim of the Prospective Observational Trial to Optimize Pediatric Health in Fetal Growth Restriction NeuroDevelopmental Assessment Study was to determine whether children born after fetal growth-restricted pregnancies are at additional risk of adverse early childhood developmental outcomes compared with children born small for gestational age. The objective of this secondary analysis was to describe the role of cerebroplacental ratio in the prediction of adverse early childhood neurodevelopmental outcome. STUDY DESIGN: Participants were recruited prospectively from the Perinatal Ireland multicenter observational Prospective Observational Trial to Optimize Pediatric Health in Fetal Growth Restriction study cohort. Fetal growth restriction was defined as birthweight <10th percentile with abnormal antenatal umbilical artery Doppler indices. Small for gestational age was defined similarly in the absence of abnormal Doppler indices. Cerebroplacental ratio was calculated with the pulsatility indices of the middle cerebral artery and divided by umbilical artery with an abnormal value <1. Children (n=375) were assessed at 3 years with the use of the Ages and Stages Questionnaire and the Bayley Scales of Infant and Toddler Development, 3rd edition. Small-for-gestational-age pregnancies with normal Doppler indices were compared with (1) fetal growth-restricted cases with abnormal umbilical artery Doppler and normal cerebroplacental ratio or (2) fetal growth restriction cases with both abnormal umbilical artery and cerebroplacental ratio. Statistical analysis was performed with statistical software via 2-sample t-test with Bonferroni adjustment, and a probability value of .00625 was considered significant. RESULTS: Assessments were performed on 198 small-for-gestational-age children, 136 fetal growth-restricted children with abnormal umbilical artery Doppler images and normal cerebroplacental ratio, and 41 fetal growth-restricted children with both abnormal umbilical artery Doppler and cerebroplacental ratio. At 3 years of age, although there were no differences in head circumference, children who also had an abnormal cerebroplacental ratio had persistently shorter stature (P=.005) and lower weight (P=.18). Children from fetal growth restriction-affected pregnancies demonstrated poorer neurodevelopmental outcome than their small-for-gestational-age counterparts. Fetal growth-restricted pregnancies with an abnormal cerebroplacental ratio had significantly poorer neurologic outcome at 3 years of age across all measured variables. CONCLUSION: We have demonstrated that growth-restricted pregnancies with a cerebroplacental ratio <1 have a significantly increased risk of delayed neurodevelopment at 3 years of age when compared with pregnancies with abnormal umbilical artery Doppler evidence alone. This study further substantiates the benefit of routine assessment of cerebroplacental ratio in fetal growth-restricted pregnancies and for counseling parents regarding the long-term outcome of affected infants.


Subject(s)
Fetal Growth Retardation/physiopathology , Middle Cerebral Artery/physiopathology , Neurodevelopmental Disorders/etiology , Pulsatile Flow , Umbilical Arteries/physiopathology , Adult , Brain/embryology , Brain/physiopathology , Child, Preschool , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Infant, Small for Gestational Age , Male , Middle Cerebral Artery/diagnostic imaging , Middle Cerebral Artery/embryology , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/physiopathology , Neuropsychological Tests , Placenta/embryology , Placenta/physiopathology , Pregnancy , Prospective Studies , Risk Factors , Ultrasonography, Doppler , Ultrasonography, Prenatal , Umbilical Arteries/diagnostic imaging , Umbilical Arteries/embryology
14.
Pharmacol Res ; 149: 104468, 2019 11.
Article in English | MEDLINE | ID: mdl-31600597

ABSTRACT

The placenta plays a critical role in mammalian reproduction. Although it is a transient organ, its function is indispensable to communication between the mother and fetus, and supply of nutrients and oxygen to the growing fetus. During pregnancy, the placenta is vulnerable to various intrinsic and extrinsic conditions which can result in increased risk of fetal neurodevelopmental disorders as well as fetal death. The placenta controls the neuroendocrine secretion in the brain as a means of adaptive processes to safeguard the fetus from adverse programs, to optimize fetal development and other physiological changes necessary for reproductive success. Although a wealth of information is available on neuroendocrine functions in pregnancy, they are largely limited to the regulation of hypothalamus-pituitary-adrenal/gonad (HPA/ HPG) axis, particularly the oxytocin and prolactin system. There is a major gap in knowledge on systems-level functional interaction between the brain and placenta. In this review, we aim to outline the current state of knowledge about the brain-placental axis with description of the functional interactions between the placenta and the maternal and fetal brain. While describing the brain-placental interactions, a special emphasis has been given on the therapeutics and pharmacology of the placental receptors to neuroligands expressed in the brain during gestation. As a key feature of this review, we outline the prospects of integrated pharmacogenomics, single-cell sequencing and organ-on-chip systems to foster priority areas in this field of research. Finally, we remark on the application of precision genomics approaches to study the brain-placental axis in order to accelerate personalized medicine and therapeutics to treat placental and fetal brain disorders.


Subject(s)
Brain/metabolism , Fetal Development/drug effects , Maternal-Fetal Exchange/physiology , Placenta/metabolism , Animals , Brain/embryology , Female , Fetal Development/genetics , Humans , Maternal-Fetal Exchange/genetics , Pharmaceutical Preparations/metabolism , Pharmacogenetics , Placenta/embryology , Pregnancy , Xenobiotics/pharmacokinetics , Xenobiotics/pharmacology
15.
Mol Biol Rep ; 46(3): 3193-3201, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30941645

ABSTRACT

Folate is an essential micronutrient during pregnancy. The differential expression of genes related to folate transport and metabolism during the advancing gestation and pregnancy complications is not well established. Hence, we studied the gene expression of folate metabolism and transport proteins in the placenta with advancing gestation, preeclampsia and neural tube defects (NTD). The expression of folate transporters and enzymes involved in folate metabolism in the placenta with advancing gestation and pregnancy-related disorders were studied by 2-step RT-PCR. Folate levels were estimated by microbiological assay using Lactobacillus casei. Significant changes in levels of placental folate metabolizing enzymes were found in both physiological and pathological pregnancies during advancing gestation. Expression of methyltetrahydrofolate reductase (MTHFR) (p < 0.001) and cystathionine-ß-synthase (CBS) (p < 0.001) was decreased while that of methionine synthase (MS) (p < 0.001) was increased with advancing gestation. A much-reduced expression of MTHFR (p < 0.01) and an abnormally high expression of methionine synthase reductase (p < 0.001) were observed in the NTD group. In NTDs, there was an adaptive up-regulation of folate transporters mainly reduced folate carrier (p < 0.001) and folate receptor alpha (p < 0.001). MTHFR expression showed a strong positive correlation (r = 0.96, p < 0.01) with folate levels in placenta. Pregnant women with preeclampsia had low expression of MS (p < 0.01) in association with low folate levels. Placental folate metabolizing enzymes exhibited a differential pattern during advancing gestation. Deficient folate status in association with alteration in expression of enzymes involved in folate metabolism might be associated with pregnancy complications such as preeclampsia and NTDs.


Subject(s)
Folic Acid/metabolism , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Organogenesis/genetics , Placenta/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Adult , Biological Transport , Female , Gene Expression Regulation , Gene Expression Regulation, Enzymologic , Humans , Metabolic Networks and Pathways , Placenta/embryology , Pre-Eclampsia/physiopathology , Pregnancy , Young Adult
16.
Pediatr Dev Pathol ; 22(6): 513-522, 2019.
Article in English | MEDLINE | ID: mdl-31138053

ABSTRACT

BACKGROUND: Placental pathology in fetal congenital anomalies in second half of pregnancy is largely unknown. METHODS: Twenty-six clinical and 45 independent placental phenotypes from pregnancies ≥20 weeks of gestation with congenital anomalies divided into 4 groups were retrospectively compared with analysis of variance or χ 2 with 3 degrees of freedom and with Bonferroni correction for multiple comparisons: group 1 : 112 cases with heart malformations (with or without chromosomal anomalies), group 2 : 41 cases with abnormal karyotypes and anomalies other than heart malformations, group 3 : 87 cases with intrathoracic or intraabdominal mass-forming anomalies (mostly congenital diaphragmatic hernias and adenomatoid airway malformation), and group 4 : 291 miscellaneous cases with mostly skeletal, renal, and central nervous system anomalies not fulfilling the criteria of inclusion into groups 1 to 3. RESULTS: Eight of 26 clinical (30.8%) and 16 of 45 (35.5%) placental phenotypes varied statistically significantly among the 4 groups (P < .05), of those, 7 (26.9%) and 4 (8.9%), respectively, remained statistically significant after Bonferroni correction (P Bonferroni ≤ .002). Those placental phenotypes were placental weight, chorionic disc chorionic microcysts, fetal vascular ectasia, and luminal vascular abnormalities of chorionic villi. CONCLUSIONS: Fetal anomalies in second half of pregnancy feature abnormal clinical phenotypes much more frequently than abnormal placental phenotypes. Chromosomal abnormalities with or without heart malformations tend to feature villous edema, and erythroblastosis of fetal blood, likely due to fetal heart failure. Mass-forming fetal anomalies feature placental histological lesions of shallow placental implantation, diffuse chronic hypoxic patterns of placental injury, and lesions of fetal vascular malperfusion, likely stasis-induced.


Subject(s)
Congenital Abnormalities/embryology , Placenta Diseases/etiology , Placenta/pathology , Female , Humans , Phenotype , Placenta/embryology , Placenta Diseases/diagnosis , Placenta Diseases/pathology , Pregnancy , Pregnancy Trimester, Second , Pregnancy Trimester, Third , Retrospective Studies
17.
Genes Dev ; 25(16): 1702-15, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21852535

ABSTRACT

The noncoding Tsix RNA is an antisense repressor of Xist and regulates X inactivation in mice. Tsix is essential for preventing the inactivation of the maternally inherited X chromosome in extraembryonic lineages where imprinted X-chromosome inactivation (XCI) occurs. Here we establish an inducible Tsix expression system for investigating Tsix function in development. We show that Tsix has a clear functional window in extraembryonic development. Within this window, Tsix can repress Xist, which is accompanied by DNA methylation of the Xist promoter. As a consequence of Xist repression, reactivation of the inactive X chromosome (Xi) is widely observed. In the parietal endoderm, Tsix represses Xist and causes reactivation of an Xi-linked GFP transgene throughout development, whereas Tsix progressively loses its Xist-repressing function from embryonic day 9.5 (E9.5) onward in trophoblast giant cells and spongiotrophoblast, suggesting that Tsix function depends on a lineage-specific environment. Our data also demonstrate that the maintenance of imprinted XCI requires Xist expression in specific extraembryonic tissues throughout development. This finding shows that reversible XCI is not exclusive to pluripotent cells, and that in some lineages cell differentiation is not accompanied by a stabilization of the Xi.


Subject(s)
RNA, Untranslated/genetics , X Chromosome Inactivation/genetics , X Chromosome/genetics , Animals , Cell Lineage/genetics , DNA Methylation , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Situ Hybridization, Fluorescence , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Placenta/cytology , Placenta/embryology , Placenta/metabolism , Pregnancy , RNA, Long Noncoding , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Trophoblasts/metabolism
18.
J Cell Mol Med ; 22(2): 716-727, 2018 02.
Article in English | MEDLINE | ID: mdl-29160594

ABSTRACT

Leptin is now considered an important signalling molecule of the reproductive system, as it regulates the production of gonadotrophins, the blastocyst formation and implantation, the normal placentation, as well as the foeto-placental communication. Leptin is a peptide hormone secreted mainly by adipose tissue, and the placenta is the second leptin-producing tissue in humans. Placental leptin is an important cytokine which regulates placental functions in an autocrine or paracrine manner. Leptin seems to play a crucial role during the first stages of pregnancy as it modulates critical processes such as proliferation, protein synthesis, invasion and apoptosis in placental cells. Furthermore, deregulation of leptin levels has been correlated with the pathogenesis of various disorders associated with reproduction and gestation, including polycystic ovary syndrome, recurrent miscarriage, gestational diabetes mellitus, pre-eclampsia and intrauterine growth restriction. Due to the relevant incidence of the mentioned diseases and the importance of leptin, we decided to review the latest information available about leptin action in normal and pathological pregnancies to support the idea of leptin as an important factor and/or predictor of diverse disorders associated with reproduction and pregnancy.


Subject(s)
Leptin/metabolism , Pregnancy Complications/metabolism , Adipose Tissue/metabolism , Female , Humans , Immunologic Factors/metabolism , Placenta/embryology , Placenta/metabolism , Pregnancy , Reproduction
19.
Dev Biol ; 432(1): 98-124, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28882402

ABSTRACT

Hypoblast/visceral endoderm assists in amniote nutrition, axial positioning and formation of the gut. Here, we provide evidence, currently limited to humans and non-human primates, that hypoblast is a purveyor of extraembryonic mesoderm in the mouse gastrula. Fate mapping a unique segment of axial extraembryonic visceral endoderm associated with the allantoic component of the primitive streak, and referred to as the "AX", revealed that visceral endoderm supplies the placentae with extraembryonic mesoderm. Exfoliation of the AX was dependent upon contact with the primitive streak, which modulated Hedgehog signaling. Resolution of the AX's epithelial-to-mesenchymal transition (EMT) by Hedgehog shaped the allantois into its characteristic projectile and individualized placental arterial vessels. A unique border cell separated the delaminating AX from the yolk sac blood islands which, situated beyond the limit of the streak, were not formed by an EMT. Over time, the AX became the hindgut lip, which contributed extensively to the posterior interface, including both embryonic and extraembryonic tissues. The AX, in turn, imparted antero-posterior (A-P) polarity on the primitive streak and promoted its elongation and differentiation into definitive endoderm. Results of heterotopic grafting supported mutually interactive functions of the AX and primitive streak, showing that together, they self-organized into a complete version of the fetal-placental interface, forming an elongated structure that exhibited A-P polarity and was composed of the allantois, an AX-derived rod-like axial extension reminiscent of the embryonic notochord, the placental arterial vasculature and visceral endoderm/hindgut.


Subject(s)
Gastrula/embryology , Placenta/embryology , Primitive Streak/cytology , Primitive Streak/embryology , Animals , Cell Differentiation/physiology , Developmental Biology/methods , Endoderm/embryology , Epithelial-Mesenchymal Transition , Female , Gastrula/metabolism , Hedgehog Proteins/metabolism , Mesoderm/embryology , Mice , Notochord/embryology , Placenta/metabolism , Pregnancy , Signal Transduction
20.
Dev Biol ; 422(2): 171-185, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27940158

ABSTRACT

A critical transition occurs near mid-gestation of mammalian pregnancy. Prior to this transition, low concentrations of oxygen (hypoxia) signaling through Hypoxia Inducible Factor (HIF) functions as a morphogen for the placenta and fetal organs. Subsequently, functional coupling of the placenta and fetal cardiovascular system for oxygen (O2) transport is required to support the continued growth and development of the fetus. Here we tested the hypothesis that Hif-1α is required in maternal cells for placental morphogenesis and function. We used Tamoxifen-inducible Cre-Lox to inactivate Hif-1α in maternal tissues at E8.5 (MATcKO), and used ODD-Luciferase as a reporter of hypoxia in placenta and fetal tissues. MATcKO of Hif-1α reduced the number of uterine natural killer (uNK) cells and Tpbpa-positve trophoblast cells in the maternal decidua at E13.5 -15.5. There were dynamic changes in all three layers of E13.5-15.5 MATcKO placenta. Of note was the under-development of the labyrinth at E15.5 associated with reduced Ki67 and increased TUNEL staining consistent with reduced cell proliferation and increased apoptosis. Labyrinth defects were particularly evident in placentas connected to effectively HIF-1α heterozygous null embryos. MATcKO had no effect on basal ODD-Luciferase activity in fetal organs (heart, liver, brain) at any stage, but at E13.5-15.5 resulted in enhanced induction of the ODD-Luciferase hypoxia reporter when the dam's inspired O2 was reduced to 8% for 4 hours. MATcKO also slowed the growth after E13.5 of fetuses that were effectively heterozygous for Hif-1α, with most being non-viable at E15.5. The hearts of these E15.5 fetuses were abnormal with reduction in size, thickened epicardium and mesenchymal septum. We conclude that maternal HIF-1α is required for placentation including recruitment of uNK and trophoblast cells into the maternal decidua and other trophoblast cell behaviors. The placental defects render the fetus vulnerable to O2 deprivation after mid-gestation.


Subject(s)
Cell Hypoxia/physiology , Heart/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Placenta/embryology , Placentation/genetics , Animals , Apoptosis , Cell Proliferation , Female , Heart/growth & development , Heart Defects, Congenital/embryology , In Situ Nick-End Labeling , Killer Cells, Natural/immunology , Mice , Oxygen/metabolism , Placenta/abnormalities , Placenta/cytology , Placentation/physiology , Pregnancy , Pregnancy Proteins/metabolism , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL