Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Prenat Diagn ; 43(2): 261-271, 2023 02.
Article En | MEDLINE | ID: mdl-36336873

Turner syndrome (TS), a common chromosomal abnormality affecting females, is associated with partial or complete loss of the second sex chromosome. Although the classic karyotype is 45, X, the detection of mosaic TS is increasing. TS is a multi-system disorder with significant endocrine, cardiovascular and reproductive impacts. Accelerated ovarian follicular loss leads to primary amenorrhoea or premature ovarian insufficiency and infertility. Early diagnosis and counselling regarding hormone replacement therapy and future reproductive capacity, including fertility preservation, are essential to improve reproductive outcomes. Pubertal induction or estrogen replacement is usually required to optimise long-term health outcomes; however, initiation may be delayed due to delayed diagnosis. Spontaneous pregnancy occurs in a small number of women; however, many require donor oocytes and assisted reproductive technology to achieve a pregnancy. Pregnancy is a high risk especially when associated with congenital heart disease. Prepregnancy counselling by the multidisciplinary team (MDT) to identify contraindications and optimise pre-existing health issues is essential. Pregnancy management should be led by a maternal-fetal medicine unit with input from the MDT. This review examines reproductive health outcomes in women with TS and how best to manage them to reduce health risks and improve maternal and neonatal outcomes.


Primary Ovarian Insufficiency , Turner Syndrome , Pregnancy , Humans , Female , Turner Syndrome/complications , Turner Syndrome/diagnosis , Turner Syndrome/genetics , Reproductive Health , Primary Ovarian Insufficiency/complications , Chromosome Aberrations , Reproductive Techniques, Assisted
2.
Sci Rep ; 11(1): 16595, 2021 08 16.
Article En | MEDLINE | ID: mdl-34400721

Fetal growth restriction is a leading cause of stillbirth that often remains undetected during pregnancy. Identifying novel biomarkers may improve detection of pregnancies at risk. This study aimed to assess syndecan-1 as a biomarker for small for gestational age (SGA) or fetal growth restricted (FGR) pregnancies and determine its molecular regulation. Circulating maternal syndecan-1 was measured in several cohorts; a large prospective cohort collected around 36 weeks' gestation (n = 1206), a case control study from the Manchester Antenatal Vascular service (285 women sampled at 24-34 weeks' gestation); two prospective cohorts collected on the day of delivery (36 + 3-41 + 3 weeks' gestation, n = 562 and n = 405 respectively) and a cohort who delivered for preterm FGR (< 34 weeks). Circulating syndecan-1 was consistently reduced in women destined to deliver growth restricted infants and those delivering for preterm disease. Syndecan-1 secretion was reduced by hypoxia, and its loss impaired proliferation. Matrix metalloproteinases and mitochondrial electron transport chain inhibitors significantly reduced syndecan-1 secretion, an effect that was rescued by coadministration of succinate, a mitochondrial electron transport chain activator. In conclusion, circulating syndecan-1 is reduced among cases of term and preterm growth restriction and has potential for inclusion in multi-marker algorithms to improve detection of poorly grown fetuses.


Fetal Growth Retardation/blood , Matrix Metalloproteinases/physiology , Mitochondria/physiology , Placenta/metabolism , Pregnancy Complications/blood , Syndecan-1/blood , Adult , Area Under Curve , Birth Weight , Cell Hypoxia , Delivery, Obstetric , Diabetes, Gestational/blood , Electron Transport/drug effects , Female , Gestational Age , Humans , Hypertension/blood , Infant, Newborn , Infant, Small for Gestational Age , Metformin/pharmacology , Mitochondria/drug effects , Organ Size , Overweight/blood , Pre-Eclampsia/blood , Pregnancy , ROC Curve , Smoking/blood , Trophoblasts/enzymology
3.
Placenta ; 114: 22-28, 2021 10.
Article En | MEDLINE | ID: mdl-34418751

INTRODUCTION: Preeclampsia is associated with reduced pro-angiogenic Placental Growth Factor (PlGF) and increased levels of anti-angiogenic soluble FMS like tyrosine kinase-1 (sFlt-1). We have previously shown that sFlt-1 secretion is positively regulated via the Epidermal Growth Factor Receptor (EGFR) and mitochondrial respiration pathways. We assessed whether these pathways also regulate endothelial and placental secretion of PlGF. METHODS: Primary cytotrophoblast cells and primary human umbilical vein endothelial cells (HUVECs) were treated with EGFR inhibitor gefitinib, or small molecules that inhibit down-stream pathways of the receptor: U0126, PD98059 (ERK/MEK pathway inhibitors), ZM336372 (JAK/STAT inhibitor) or AG490 (JAK inhibitor). We inhibited mitochondrial respiration in primary cytotrophoblasts using mitochondrial complex inhibitors rotenone (complex I), antimycin (complex III) or oligomycin (complex IV). We then measured PlGF secretion in the condition media. RESULTS: Three inhibitors of the EGFR pathway significantly increased PlGF secretion: gefitinib (p = 0.03), AG490 (p < 0.0001) and U0126 (p = 0.03) in primary cytotrophoblasts, while PD98059 reduced PlGF secretion (p = 0.002). In the same cells, neither gefitinib or UO126 altered PlGF mRNA expression, but AG490 significantly increased its expression (p = 0.02). Primary endothelial cell PlGF secretion was significantly reduced when treated with PD98059 and U0126 while ZM336372 had no effect. Rotenone significantly reduced cytotrophoblast PlGF secretion (p = 0.0005). Neither antimycin (p = 0.9) or oligomycin (p = 0.9) had an effect. DISCUSSION: We have shown that PlGF secretion from primary cytotrophoblast and HUVECs is altered by inhibiting EGFR signaling and potentially mitochondrial respiration, coincident with reduced sFlt-1 secretion. This suggests that common pathways are regulating both pro and anti-angiogenic molecules that are changed in association with preeclampsia and provides insight into the pathogenesis of this serious disease.


Epidermal Growth Factor/metabolism , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/metabolism , Placenta Growth Factor/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , Signal Transduction/genetics , Adult , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/genetics , Female , Gefitinib/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Placenta/drug effects , Placenta Growth Factor/genetics , Pregnancy , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Trophoblasts/metabolism
4.
Int J Mol Sci ; 22(14)2021 Jul 12.
Article En | MEDLINE | ID: mdl-34299087

Biomarkers for placental dysfunction are currently lacking. We recently identified SPINT1 as a novel biomarker; SPINT2 is a functionally related placental protease inhibitor. This study aimed to characterise SPINT2 expression in placental insufficiency. Circulating SPINT2 was assessed in three prospective cohorts, collected at the following: (1) term delivery (n = 227), (2) 36 weeks (n = 364), and (3) 24-34 weeks' (n = 294) gestation. SPINT2 was also measured in the plasma and placentas of women with established placental disease at preterm (<34 weeks) delivery. Using first-trimester human trophoblast stem cells, SPINT2 expression was assessed in hypoxia/normoxia (1% vs. 8% O2), and following inflammatory cytokine treatment (TNFα, IL-6). Placental SPINT2 mRNA was measured in a rat model of late-gestational foetal growth restriction. At 36 weeks, circulating SPINT2 was elevated in patients who later developed preeclampsia (p = 0.028; median = 2233 pg/mL vs. controls, median = 1644 pg/mL), or delivered a small-for-gestational-age infant (p = 0.002; median = 2109 pg/mL vs. controls, median = 1614 pg/mL). SPINT2 was elevated in the placentas of patients who required delivery for preterm preeclampsia (p = 0.025). Though inflammatory cytokines had no effect, hypoxia increased SPINT2 in cytotrophoblast stem cells, and its expression was elevated in the placental labyrinth of growth-restricted rats. These findings suggest elevated SPINT2 is associated with placental insufficiency.


Biomarkers/metabolism , Fetal Growth Retardation/diagnosis , Membrane Glycoproteins/metabolism , Placenta Diseases/diagnosis , Placenta/pathology , Pre-Eclampsia/diagnosis , Trophoblasts/pathology , Adolescent , Female , Fetal Growth Retardation/metabolism , Humans , Infant, Newborn , Infant, Small for Gestational Age , Longitudinal Studies , Placenta/metabolism , Placenta Diseases/metabolism , Pre-Eclampsia/metabolism , Pregnancy , Prospective Studies , Trophoblasts/metabolism
5.
Sci Rep ; 10(1): 18077, 2020 10 22.
Article En | MEDLINE | ID: mdl-33093531

Preeclampsia is a pregnancy complication associated with angiogenic dysbalance, maternal endothelial dysfunction and end-organ injury. A predictive test to identify those who will develop preeclampsia could substantially decrease morbidity and mortality. MicroRNAs (miRs) are small RNA molecules involved in post-transcriptional gene regulation. We screened for circulating miRs differentially expressed at 36 weeks' gestation in pregnancies before the development of preeclampsia. We used a case-control group (198 controls, 34 pre-preeclampsia diagnosis) selected from a prospective cohort (n = 2015) and performed a PCR-based microarray to measure the expression of 41 miRs. We found six circulating miRs (miRs 363, 149, 18a, 1283, 16, 424) at 36 weeks' had significantly reduced expression (p < 0.0001-0.04). miR363 was significantly downregulated at 28 weeks' gestation, 10-12 weeks before the onset of clinical disease. In the circulation of another cohort of 34 participants with established preterm preeclampsia (vs 23 controls), we found miRs363, 18a, 149 and 16 were significantly down regulated (p < 0.0001-0.04). Combined expression of miRs149 and 363 in the circulation at 36 weeks' gestation provides a test with 45% sensitivity (at a specificity of 90%) which suggests measuring both miRs may have promise as part of a multi-marker test to predict preeclampsia.


Biomarkers/blood , MicroRNAs/blood , Pre-Eclampsia/diagnosis , Case-Control Studies , Female , Follow-Up Studies , Gestational Age , Humans , Infant, Newborn , Microarray Analysis , Pre-Eclampsia/blood , Pre-Eclampsia/genetics , Pregnancy , Prognosis , Prospective Studies
6.
Pregnancy Hypertens ; 16: 16-25, 2019 Apr.
Article En | MEDLINE | ID: mdl-31056153

Preeclampsia is a pregnancy complication associated with elevated placental secretion of anti-angiogenic factors, maternal endothelial dysfunction and end-organ injury. Adrenomedullin (ADM) is a pro-angiogenic peptide hormone which regulates blood pressure and vascular integrity. It is highly expressed in both the placenta and vascular endothelial cells. We performed a nested case-control study, selected from a large prospective cohort of over 2000 participants. Circulating ADM mRNA was reduced at both 28 (n = 39 vs 248 controls, p = 0.005) and 36 weeks' of pregnancy (n = 39 vs 205 controls, p < 0.0001) in those destined to develop term preeclampsia. It was also significantly reduced in the circulation of women with established early-onset preeclampsia (n = 34 vs 21 controls, p = 0.01). ADM mRNA (n = 34 vs 12 controls) and protein (n = 53 vs 17 controls) were significantly decreased in placental tissue from women with early-onset preeclampsia (p = 0.02, p = 0.0002 respectively), suggesting the placenta is a possible source of the reduced circulating mRNA. Functional studies in primary endothelial cells revealed significantly reduced ADM mRNA expression when cells were exposed to cytotrophoblast conditioned media (derived from normotensive pregnancies, p < 0.0001) or TNFα (p < 0.0001), suggesting another possible source of reduced circulating ADM mRNA is the endothelium. Circulating ADM mRNA, but not protein, is reduced 10-12 weeks before the diagnosis of term preeclampsia. It may be of endothelial or placental origin. Whole blood mRNA is a rich source of potential biomarker discovery in the prediction of preeclampsia.


Adrenomedullin/genetics , Pre-Eclampsia/diagnosis , Prenatal Diagnosis , RNA, Messenger/blood , Adult , Biomarkers/blood , Case-Control Studies , Cell-Free Nucleic Acids/blood , Cohort Studies , Female , Humans , Pre-Eclampsia/blood , Pregnancy , Pregnancy Trimester, Third , Prospective Studies , Sensitivity and Specificity
7.
Placenta ; 84: 28-31, 2019 09 01.
Article En | MEDLINE | ID: mdl-30745114

Pregnancy complications such as fetal growth restriction and preeclampsia are diseases with limited biomarkers for prediction, and a complete lack of therapeutic options. We define placenta-enriched molecules as those that are highly expressed in the placenta relative to all other human tissues. Many exist including mRNAs, miRNAs and proteins. It is now well established that placenta-enriched mRNAs are found within the maternal circulation and are cleared rapidly after birth. Similarly, distinct clusters of miRNAs that are placenta-enriched have been identified and are measurable within the circulation. However, perhaps the most established potential diagnostics thus far are circulating placental proteins such as placental growth factor (PlGF), pregnancy associated pregnancy protein-A (PAPP-A) and soluble FMS-like tyrosine kinase 1 (sFlt-1). There has also been much interest in targeting placenta-enriched molecules as a means to treat diseases of pregnancy. We have shown promising results in targeting placenta-enriched epidermal growth factor receptor (EGFR) to treat ectopic pregnancy. Others have focused on using placenta-enriched molecules as a means of homing therapeutic-filled nanoparticles to the placenta, or to directly target sFlt-1 to improve disease outcomes. Importantly, many placenta-enriched molecules remain largely unstudied. We propose that a better understanding of their biology, and potential contribution to the pathogenesis of diseases, may yield more predictive diagnostic and therapeutic targets.


Biomarkers/metabolism , Molecular Diagnostic Techniques/methods , Pregnancy Complications/therapy , Pregnancy Proteins/physiology , Prenatal Diagnosis/methods , Biomarkers/analysis , Drug Development/methods , Drug Development/trends , Female , Humans , MicroRNAs/physiology , Placenta/metabolism , Pregnancy , Pregnancy Complications/diagnosis
8.
Sci Rep ; 9(1): 235, 2019 01 18.
Article En | MEDLINE | ID: mdl-30659233

Preeclampsia is a pregnancy complication associated with elevated placental secretion of anti-angiogenic factors, maternal endothelial dysfunction and organ injury. GATA2 is a transcription factor expressed in the endothelium which regulates vascular homeostasis by controlling transcription of genes and microRNAs, including endothelial miR126. We assessed GATA2 and miR126 in preeclampsia. Whole blood circulating GATA2 mRNA and miR126 expression were significantly decreased in women with established early-onset preeclampsia compared to gestation-matched controls (p = 0.002, p < 0.0001, respectively). Using case-control groups selected from a large prospective cohort, whole blood circulating GATA2 mRNA at both 28 and 36 weeks' gestation was significantly reduced prior to the clinical diagnosis of preeclampsia (p = 0.012, p = 0.015 respectively). There were no differences in GATA2 mRNA or protein expression in preeclamptic placentas compared to controls, suggesting the placenta is an unlikely source. Inducing endothelial dysfunction in vitro by administering either tumour necrosis factor-α or placenta-conditioned media to endothelial cells, significantly reduced GATA2 mRNA expression (p < 0.0001), suggesting the reduced levels of circulating GATA2 mRNA may be of endothelial origin. Circulating GATA2 mRNA is decreased in women with established preeclampsia and decreased up to 12 weeks preceding onset of disease. Circulating mRNAs of endothelial origin may be a novel source of biomarker discovery for preeclampsia.


Biomarkers/blood , Endothelial Cells/metabolism , GATA2 Transcription Factor/genetics , Pre-Eclampsia/diagnosis , RNA, Messenger/blood , Adult , Case-Control Studies , Female , GATA2 Transcription Factor/biosynthesis , Humans , MicroRNAs/biosynthesis , Pregnancy , Prospective Studies
9.
J Matern Fetal Neonatal Med ; 29(23): 3866-9, 2016 Dec.
Article En | MEDLINE | ID: mdl-26940081

OBJECTIVE: We compared the efficacy of Carbetocin (long-acting oxytocin receptor agonist) versus Oxytocin given at non-elective caesarean section. METHOD: We performed a double-blind, randomised, single-centre study. Eligible women were ≥37 weeks of gestation undergoing emergency caesarean section. Participants received either carbetocin of 100 µg or oxytocin 5 international units. The primary outcome was the need to administer additional uterotonics, as determined by the clinician. Secondary outcomes included estimated blood loss, haemoglobin drop pre-post operation and the need for a blood transfusion Results: From August 2012 to February 2013, 114 women were enroled. Two were excluded from analysis as they received a general anaesthetic. Fifty-nine patients received 100-µg carbetocin; 53 received 5 international units oxytocin. There was no statistically significant difference in the number of women requiring additional uterotonics between the two groups: Carbetocin group 22% and Oxytocin group 13% (p = 0.323). There were no significant differences in the fall in haemoglobin, estimated blood loss, rates of post-partum haemorrhage or blood transfusions. CONCLUSION: Oxytocin and carbetocin have similar requirements for additional uterotonics, estimated blood loss, haemoglobin drop and blood transfusions. There was a trend towards requiring additional uterotonics in patients receiving Carbetocin which was not statistically significant. This study found no benefits in using carbetocin over oxytocin.


Cesarean Section/methods , Oxytocics/administration & dosage , Oxytocin/analogs & derivatives , Oxytocin/administration & dosage , Postpartum Hemorrhage/prevention & control , Adult , Double-Blind Method , Female , Humans , Pregnancy , Young Adult
...