1
|
Hinchliffe E, Heazell A. Profiling neuroactive steroids in pregnancy. A non-derivatised liquid chromatography tandem mass spectrometry method for the quantitation of allopregnanolone and four related isomers in maternal serum. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1256:124541. [PMID: 40054418 DOI: 10.1016/j.jchromb.2025.124541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/28/2025] [Accepted: 02/23/2025] [Indexed: 04/07/2025]
Abstract
Neuroactive steroids are metabolites of progesterone, synthesised during pregnancy by the placenta. Here, we describe development of a novel liquid chromatography tandem mass spectrometry (LC-MS/MS) assay for quantitation of allopregnanolone, pregnanolone, isopregnanolone, epipregnanolone and allopregnan-20α-ol-3-one in maternal serum. Following addition of deuterated internal standards, 200 μL of serum was subjected to solid phase extraction. Chromatography was performed using a pentafluorophenyl column, and LC-MS/MS on a Sciex 6500+. Sample injection volume was 20 μL, and injection-to-injection time 10.0 min. The assay was validated according to published guidelines; assay linearity and lower limit of quantification were suitable for analysis of each steroid in maternal serum, for all analytes mean recoveries were 100 % ± 15 %, intra- and inter-assay imprecision <15 %, and matrix effects negligible, and specificity experiments confirmed nil interference from a wide range of endogenous metabolites of progesterone. The method was applied to human serum samples obtained from a large cohort of third trimester pregnancies which were subsequently characterised by normal fetal and maternal outcomes, and relationships between maternal neuroactive steroid concentrations and fetal gestational age assessed. Positive correlations between maternal serum concentration and fetal gestational age were observed for isopregnanolone, allopregnanolone and allopregnan-20α-ol-3-one. The LC-MS/MS method offers significant advantages over previously published approaches for quantitation of neuroactive steroids in human maternal serum, notably obviating the need for derivatisation, whilst achieving exceptional specificity. Characterisation of normal maternal neuroactive steroid concentrations will aid future research as dysregulated placental progesterone metabolism is observed in pregnancies with poor outcomes.
Collapse
Affiliation(s)
- Edward Hinchliffe
- Dept Clinical Biochemistry, Manchester University NHS Foundation Trust, Manchester, UK.
| | - Alexander Heazell
- Maternal and Fetal Health Research Centre, School of Biomedicine, University of Manchester, Manchester, UK; Department of Obstetrics, Saint Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
2
|
Tian Y, Liu M, Sun JY, Wang Y, Chen L, Sun W, Zhou L. Diagnosis of preeclampsia using metabolomic biomarkers. Hypertens Pregnancy 2024; 43:2379386. [PMID: 39039822 DOI: 10.1080/10641955.2024.2379386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/30/2024] [Indexed: 07/24/2024]
Abstract
The diagnostic criteria for preeclampsia do not accurately reflect the pathophysiological characteristics of patients with preeclampsia. Conventional biomarkers and diagnostic approaches have proven insufficient to fully comprehend the intricacies of preeclampsia. This study aimed to screen differentially abundant metabolites as candidate biomarkers for preeclampsia. A propensity score matching method was used to perform a 1:1 match between preeclampsia patients (n = 70) and healthy control individuals (n = 70). Based on univariate and multivariate statistical analysis methods, the different characteristic metabolites were screened and identified. Least absolute shrinkage and selection operator (LASSO) regression analysis was subsequently used to further screen for differentially abundant metabolites. A receiver operating characteristic (ROC) curve was drawn to evaluate the diagnostic efficacy of the metabolites. A total of 1,630 metabolites were identified and quantified in maternal serum samples. Fifty-three metabolites were significantly increased, and two were significantly decreased in preeclampsia patients. The area under the curve (AUC) of the model composed of isobutyryl-L-carnitine and acetyl-leucine was 0.878, and the sensitivity and specificity in detecting preeclampsia were 81.4% and 87.1%, respectively. There are significant differences in metabolism between preeclampsia patients and healthy pregnant women, and a range of novel biomarkers have been identified. These findings lay the foundation for the use of metabolomic biomarkers for the diagnosis of preeclampsia.
Collapse
Affiliation(s)
- Yunfan Tian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mingwei Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jin-Yu Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yifeng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lianmin Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ling Zhou
- Department of Obstetrics and Gynecology, Liyang People's Hospital, Liyang, Jiangsu, China
| |
Collapse
|
3
|
Adamo KB, Goudreau AD, Corson AE, MacDonald ML, O'Rourke N, Tzaneva V. Physically active pregnancies: Insights from the placenta. Physiol Rep 2024; 12:e16104. [PMID: 38872466 PMCID: PMC11176744 DOI: 10.14814/phy2.16104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/03/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Abstract
Physical activity (PA) positively influences pregnancy, a critical period for health promotion, and affects placental structure and function in ways previously overlooked. Here, we summarize the current body of literature examining the association between PA, placenta biology, and physiology while also highlighting areas where gaps in knowledge exist. PA during pregnancy induces metabolic changes, influencing nutrient availability and transporter expression in the placenta. Hormones and cytokines secreted during PA contribute to health benefits, with intricate interactions in pro- and anti-inflammatory markers. Extracellular vesicles and placental "-omics" data suggest that gestational PA can shape placental biology, affecting gene expression, DNA methylation, metabolite profiles, and protein regulation. However, whether cytokines that respond to PA alter placental proteomic profiles during pregnancy remains to be elucidated. The limited research on placenta mitochondria of physically active gestational parents (gesP), has shown improvements in mitochondrial DNA and antioxidant capacity, but the relationship between PA, placental mitochondrial dynamics, and lipid metabolism remains unexplored. Additionally, PA influences the placenta-immune microenvironment, angiogenesis, and may confer positive effects on neurodevelopment and mental health through placental changes, vascularization, and modulation of brain-derived neurotrophic factor. Ongoing exploration is crucial for unraveling the multifaceted impact of PA on the intricate placental environment.
Collapse
Affiliation(s)
- Kristi B Adamo
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Alexandra D Goudreau
- Department of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Abbey E Corson
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Meaghan L MacDonald
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Nicholas O'Rourke
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Velislava Tzaneva
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
4
|
Wang Q, Xiong Z, Wang B, Wang W, Zheng H. Ferroptosis and Preeclampsia: Genetic Analysis of Potential Biomarkers and Therapeutic Targets. Biochem Genet 2024; 62:853-875. [PMID: 37474873 DOI: 10.1007/s10528-023-10449-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/05/2023] [Indexed: 07/22/2023]
Abstract
Ferroptosis is the oxidative death of cells attributed to an imbalance in intracellular lipid reactive oxygen species metabolism, a reduction in cell antioxidant capacity, and an accumulation of membrane lipid peroxides. Trophoblast cells are a group of cells susceptible to ferroptosis. The ferroptosis of trophoblast cells has a major effect on the development of preeclampsia (PE), although the impact of ferroptosis-related genes (FRGs) on PE has not been well characterized. This study obtained PE-related information from the Gene Expression Omnibus database and FRGs from the FerrDb ferroptosis database. Seventeen PE-related differentially expressed ferroptosis-related genes (DE-FRGs) that were closely associated with cellular regulation and immune response were obtained. According to the results of a subsequent functional enrichment analysis, it was found that the above marker genes may impact PE by regulating immune response, amino acid metabolism, the cell cycle, and multiple pathways correlated with PE pathogenesis. Subsequently, we used LASSO and support vector machine recursive feature elimination algorithms to help identify GOT1, CFL1, FZD7, VDR, PARP6, TMSB4X, VCP, and ENO3 as marker genes from the 17 obtained genes. According to the results of single-sample gene set enrichment analysis (ssGSEA), the immune microenvironment of PE changed, possibly due to the GOT1 and TMSB4X genes. Furthermore, 23 drugs targeting one marker gene were determined. A constructed ceRNA network revealed a complicated regulatory link based on the eight marker genes. In this study, diagnostic potency was developed, and insight into the mechanism of PE was provided. In-depth research should be conducted before clinical application to evaluate the diagnostic value of DE-FRGs in PE.
Collapse
Affiliation(s)
- Qingmin Wang
- Department of Obstetrics, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China
| | - Zhihui Xiong
- Department of Obstetrics, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China
| | - Baimiao Wang
- Department of Obstetrics, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China
| | - Wei Wang
- Department of Obstetrics, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China.
| | - Huiling Zheng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Xinhua Hospital of Zhejiang Province, Hangzhou, 310005, China.
| |
Collapse
|
5
|
Hartvigsson O, Barman M, Rabe H, Sandin A, Wold AE, Brunius C, Sandberg AS. Associations of the placental metabolome with immune maturation up to one year of age in the Swedish NICE-cohort. Metabolomics 2024; 20:28. [PMID: 38407648 PMCID: PMC10896773 DOI: 10.1007/s11306-024-02092-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 01/26/2024] [Indexed: 02/27/2024]
Abstract
INTRODUCTION Allergies and other immune-mediated diseases are thought to result from incomplete maturation of the immune system early in life. We previously showed that infants' metabolites at birth were associated with immune cell subtypes during infancy. The placenta supplies the fetus with nutrients, but may also provide immune maturation signals. OBJECTIVES To examine the relationship between metabolites in placental villous tissue and immune maturation during the first year of life and infant and maternal characteristics (gestational length, birth weight, sex, parity, maternal age, and BMI). METHODS Untargeted metabolomics was measured using Liquid Chromatography-Mass Spectrometry. Subpopulations of T and B cells were measured using flow cytometry at birth, 48 h, one, four, and 12 months. Random forest analysis was used to link the metabolomics data with the T and B cell sub populations as well as infant and maternal characteristics. RESULTS Modest associations (Q2 = 0.2-0.3) were found between the placental metabolome and kappa-deleting recombination excision circles (KREC) at birth and naïve B cells and memory T cells at 12 months. Weak associations were observed between the placental metabolome and sex and parity. Still, most metabolite features of interest were of low intensity compared to associations previously found in cord blood, suggesting that underlying metabolites were not of placental origin. CONCLUSION Our results indicate that metabolomic measurements of the placenta may not effectively recognize metabolites important for immune maturation.
Collapse
Affiliation(s)
- Olle Hartvigsson
- Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Göteborg, Sweden
| | - Malin Barman
- Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Göteborg, Sweden.
| | - Hardis Rabe
- Institute of Biomedicine, Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Sandin
- Department of Clinical Sciences, Unit of Pediatrics, Umeå University, Umeå, Sweden
| | - Agnes E Wold
- Institute of Biomedicine, Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carl Brunius
- Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Göteborg, Sweden
| | - Ann-Sofie Sandberg
- Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Göteborg, Sweden
| |
Collapse
|
6
|
van Zundert SKM, Broekhuizen M, Smit AJP, van Rossem L, Mirzaian M, Willemsen SP, Danser AHJ, De Rijke YB, Reiss IKM, Merkus D, Steegers-Theunissen RPM. The Role of the Kynurenine Pathway in the (Patho) physiology of Maternal Pregnancy and Fetal Outcomes: A Systematic Review. Int J Tryptophan Res 2022; 15:11786469221135545. [PMID: 36467775 PMCID: PMC9716456 DOI: 10.1177/11786469221135545] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 10/10/2022] [Indexed: 08/04/2023] Open
Abstract
INTRODUCTION Tryptophan is the precursor of kynurenine pathway (KP) metabolites which regulate immune tolerance, energy metabolism, and vascular tone. Since these processes are important during pregnancy, changes in KP metabolite concentrations may play a role in the pathophysiology of pregnancy complications. We hypothesize that KP metabolites can serve as novel biomarkers and preventive therapeutic targets. This review aimed to provide more insight into associations between KP metabolite concentrations in maternal and fetal blood, and in the placenta, and adverse maternal pregnancy and fetal outcomes. METHODS A systematic search was performed on 18 February 2022 comprising all KP metabolites, and keywords related to maternal pregnancy and fetal outcomes. English-written human studies measuring KP metabolite(s) in maternal or fetal blood or in the placenta in relation to pregnancy complications, were included. Methodological quality was assessed using the ErasmusAGE quality score (QS) (range: 0-10). A meta-analysis of the mean maternal tryptophan and kynurenine concentrations in uncomplicated pregnancies was conducted. RESULTS Of the 6262 unique records, 37 were included (median QS = 5). Tryptophan was investigated in most studies, followed by kynurenine, predominantly in maternal blood (n = 28/37), and in the second and third trimester of pregnancy (n = 29/37). Compared to uncomplicated pregnancies, decreased tryptophan in maternal blood was associated with an increased prevalence of depression, gestational diabetes mellitus, fetal growth restriction, spontaneous abortion, and preterm birth. Elevated tryptophan was only observed in women with pregnancy-induced hypertension compared to normotensive pregnant women. In women with preeclampsia, only kynurenic acid was altered; elevated in the first trimester of pregnancy, and positively associated with proteinuria in the third trimester of pregnancy. CONCLUSIONS KP metabolite concentrations were altered in a variety of maternal pregnancy and fetal complications. This review implies that physiological pregnancy requires a tight balance of KP metabolites, and that disturbances in either direction are associated with adverse maternal pregnancy and fetal outcomes.
Collapse
Affiliation(s)
- Sofie KM van Zundert
- Department of Obstetrics and
Gynecology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Clinical Chemistry,
Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Michelle Broekhuizen
- Division of Neonatology, Department of
Pediatrics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Division of Pharmacology and Vascular
Medicine, Department of Internal Medicine, Erasmus MC University Medical Center,
Rotterdam, The Netherlands
- Division of Experimental Cardiology,
Department of Cardiology, Erasmus MC University Medical Center, Rotterdam, The
Netherlands
| | - Ashley JP Smit
- Department of Obstetrics and
Gynecology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Lenie van Rossem
- Department of Obstetrics and
Gynecology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Mina Mirzaian
- Department of Clinical Chemistry,
Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Sten P Willemsen
- Department of Obstetrics and
Gynecology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Biostatistics, Erasmus MC
University Medical Center, Rotterdam, The Netherlands
| | - AH Jan Danser
- Division of Pharmacology and Vascular
Medicine, Department of Internal Medicine, Erasmus MC University Medical Center,
Rotterdam, The Netherlands
| | - Yolanda B De Rijke
- Department of Clinical Chemistry,
Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Irwin KM Reiss
- Division of Neonatology, Department of
Pediatrics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology,
Department of Cardiology, Erasmus MC University Medical Center, Rotterdam, The
Netherlands
- Walter Brendel Center of Experimental
Medicine, University Clinic Munich, Ludwig Maximillian University Munich, Munich,
Germany
| | | |
Collapse
|
7
|
Coenzyme A Restriction as a Factor Underlying Pre-Eclampsia with Polycystic Ovary Syndrome as a Risk Factor. Int J Mol Sci 2022; 23:ijms23052785. [PMID: 35269927 PMCID: PMC8911031 DOI: 10.3390/ijms23052785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/24/2022] [Indexed: 02/07/2023] Open
Abstract
Pre-eclampsia is the most common pregnancy complication affecting 1 in 20 pregnancies, characterized by high blood pressure and signs of organ damage, most often to the liver and kidneys. Metabolic network analysis of published lipidomic data points to a shortage of Coenzyme A (CoA). Gene expression profile data reveal alterations to many areas of metabolism and, crucially, to conflicting cellular regulatory mechanisms arising from the overproduction of signalling lipids driven by CoA limitation. Adverse feedback loops appear, forming sphingosine-1-phosphate (a cause of hypertension, hypoxia and inflammation), cytotoxic isoketovaleric acid (inducing acidosis and organ damage) and a thrombogenic lysophosphatidyl serine. These also induce mitochondrial and oxidative stress, leading to untimely apoptosis, which is possibly the cause of CoA restriction. This work provides a molecular basis for the signs of pre-eclampsia, why polycystic ovary syndrome is a risk factor and what might be done to treat and reduce the risk of disease.
Collapse
|
8
|
Alterations in the metabolic status of amino acids in newborns of pre-eclampsia women. Pregnancy Hypertens 2022; 27:170-172. [DOI: 10.1016/j.preghy.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 11/30/2022]
|
9
|
Cui C, Wu C, Wang J, Zheng X, Ma Z, Zhu P, Guan W, Zhang S, Chen F. Leucine supplementation during late gestation globally alters placental metabolism and nutrient transport via modulation of the PI3K/AKT/mTOR signaling pathway in sows. Food Funct 2022; 13:2083-2097. [PMID: 35107470 DOI: 10.1039/d1fo04082k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In a previously published study we reported that sow dietary leucine supplementation during late pregnancy significantly improved newborn piglet birth weight by stimulating protein synthesis in the longissimus dorsi muscle. However, there is still limited knowledge as to whether leucine can exert its effects on the placenta, one of the most important temporal organs during pregnancy, to promote maternal-fetal nutrient supply and thus contribute to fetal intrauterine development. Therefore, we tested this hypothesis in the present study. In total, 150 sows at day 90 of gestation were divided into three groups and fed with either a control diet (CON), CON + 0.4% Leu or CON + 0.8% Leu, respectively, until parturition. Placental metabolomics, full spectrum amino acids and nutrient transporters were systematically analyzed after sample collection. The results indicated that Leu supplementation led to an altered placental metabolism with an increased number of metabolites related to glycolysis and the oxidation of fatty acids, as well as elevated levels of amino acid accumulation in the placenta. In addition, nutrient transporters of amino acids, glucose and fatty acids in the placenta were globally up-regulated and several enzymes related to energy metabolism, including hexokinase, succinate dehydrogenase, lactated hydrogenase, glycogen phosphorylase and hydroxyacyl-CoA-dehydrogenase, were also significantly increased with no change observed in the antioxidative status of those groups with Leu supplementation. Furthermore, the phosphorylation of PI3K, Akt, and mTOR was enhanced in the placenta of sows undergoing Leu treatment. Collectively, we concluded that supplementing the diets of sows with Leu during late gestation globally altered placental metabolism and promoted maternal-fetus nutrient transport (amino acids, glucose, and fatty acids) via modulation of the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Chang Cui
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Caichi Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Jun Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Xiaoyu Zheng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Ziwei Ma
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Pengwei Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Wutai Guan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China. .,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Shihai Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China. .,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Fang Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China. .,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
10
|
Broekhuizen M, Danser AHJ, Reiss IKM, Merkus D. The Function of the Kynurenine Pathway in the Placenta: A Novel Pharmacotherapeutic Target? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182111545. [PMID: 34770059 PMCID: PMC8582682 DOI: 10.3390/ijerph182111545] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/26/2021] [Accepted: 10/30/2021] [Indexed: 12/21/2022]
Abstract
(L-)tryptophan is metabolized via the kynurenine pathway into several kynurenine metabolites with distinct functions. Dysfunction of the kynurenine pathway can lead to impairments in vascular regulation, immune regulation, and tolerance. The first and rate limiting enzyme of this pathway, indoleamine 2,3-dioxygenase (IDO), is highly expressed in the placenta and reduced in placentas from complicated pregnancies. IDO is essential during pregnancy, as IDO inhibition in pregnant mice resulted in fetal loss. However, the exact function of placental IDO, as well as its exact placental localization, remain controversial. This review identified that two isoforms of IDO; IDO1 and IDO2, are differently expressed between placental cells, suggesting spatial segregation. Furthermore, this review summarizes how the placental kynurenine pathway is altered in pregnancy complications, including recurrent miscarriage, preterm birth, preeclampsia, and fetal growth restriction. Importantly, we describe that these alterations do not affect maternally circulating metabolite concentrations, suggesting that the kynurenine pathway functions as a local signaling pathway. In the placenta, it is an important source of de novo placental NAD+ synthesis and regulates fetal tryptophan and kynurenine metabolite supply. Therefore, kynurenine pathway interventions might provide opportunities to treat pregnancy complications, and this review discusses how such treatment could affect placental function and pregnancy development.
Collapse
Affiliation(s)
- Michelle Broekhuizen
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
- Division of Neonatology, Department of Pediatrics, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
- Division of Experimental Cardiology, Department of Cardiology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
- Correspondence:
| | - A. H. Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Irwin K. M. Reiss
- Division of Neonatology, Department of Pediatrics, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
- Walter Brendel Center of Experimental Medicine, University Clinic Munich, LMU Munich, 81377 Munich, Germany
| |
Collapse
|
11
|
Mohammad S, Bhattacharjee J, Vasanthan T, Harris CS, Bainbridge SA, Adamo KB. Metabolomics to understand placental biology: Where are we now? Tissue Cell 2021; 73:101663. [PMID: 34653888 DOI: 10.1016/j.tice.2021.101663] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 12/16/2022]
Abstract
Metabolomics, the application of analytical chemistry methodologies to survey the chemical composition of a biological system, is used to globally profile and compare metabolites in one or more groups of samples. Given that metabolites are the terminal end-products of cellular metabolic processes, or 'phenotype' of a cell, tissue, or organism, metabolomics is valuable to the study of the maternal-fetal interface as it has the potential to reveal nuanced complexities of a biological system as well as differences over time or between individuals. The placenta acts as the primary site of maternal-fetal exchange, the success of which is paramount to growth and development of offspring during pregnancy and beyond. Although the study of metabolomics has proven moderately useful for the screening, diagnosis, and understanding of the pathophysiology of pregnancy complications, the placental metabolome in the context of a healthy pregnancy remains poorly characterized and understood. Herein, we discuss the technical aspects of metabolomics and review the current literature describing the placental metabolome in human and animal models, in the context of health and disease. Finally, we highlight areas for future opportunities in the emerging field of placental metabolomics.
Collapse
Affiliation(s)
- S Mohammad
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - J Bhattacharjee
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - T Vasanthan
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - C S Harris
- Department of Biology & Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada
| | - S A Bainbridge
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, ON, Canada
| | - K B Adamo
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
12
|
Monni G, Atzori L, Corda V, Dessolis F, Iuculano A, Hurt KJ, Murgia F. Metabolomics in Prenatal Medicine: A Review. Front Med (Lausanne) 2021; 8:645118. [PMID: 34249959 PMCID: PMC8267865 DOI: 10.3389/fmed.2021.645118] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 05/04/2021] [Indexed: 11/13/2022] Open
Abstract
Pregnancy is a complicated and insidious state with various aspects to consider, including the well-being of the mother and child. Developing better non-invasive tests that cover a broader range of disorders with lower false-positive rates is a fundamental necessity in the prenatal medicine field, and, in this sense, the application of metabolomics could be extremely useful. Metabolomics measures and analyses the products of cellular biochemistry. As a biomarker discovery tool, the integrated holistic approach of metabolomics can yield new diagnostic or therapeutic approaches. In this review, we identify and summarize prenatal metabolomics studies and identify themes and controversies. We conducted a comprehensive search of PubMed and Google Scholar for all publications through January 2020 using combinations of the following keywords: nuclear magnetic resonance, mass spectrometry, metabolic profiling, prenatal diagnosis, pregnancy, chromosomal or aneuploidy, pre-eclampsia, fetal growth restriction, pre-term labor, and congenital defect. Metabolite detection with high throughput systems aided by advanced bioinformatics and network analysis allowed for the identification of new potential prenatal biomarkers and therapeutic targets. We took into consideration the scientific papers issued between the years 2000-2020, thus observing that the larger number of them were mainly published in the last 10 years. Initial small metabolomics studies in perinatology suggest that previously unidentified biochemical pathways and predictive biomarkers may be clinically useful. Although the scientific community is considering metabolomics with increasing attention for the study of prenatal medicine as well, more in-depth studies would be useful in order to advance toward the clinic world as the obtained results appear to be still preliminary. Employing metabolomics approaches to understand fetal and perinatal pathophysiology requires further research with larger sample sizes and rigorous testing of pilot studies using various omics and traditional hypothesis-driven experimental approaches.
Collapse
Affiliation(s)
- Giovanni Monni
- Department of Prenatal and Preimplantation Genetic Diagnosis and Fetal Therapy, Ospedale Pediatrico Microcitemico “A.Cao,”Cagliari, Italy
| | - Luigi Atzori
- Clinical Metabolomics Unit, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Valentina Corda
- Department of Prenatal and Preimplantation Genetic Diagnosis and Fetal Therapy, Ospedale Pediatrico Microcitemico “A.Cao,”Cagliari, Italy
| | - Francesca Dessolis
- Department of Prenatal and Preimplantation Genetic Diagnosis and Fetal Therapy, Ospedale Pediatrico Microcitemico “A.Cao,”Cagliari, Italy
| | - Ambra Iuculano
- Department of Prenatal and Preimplantation Genetic Diagnosis and Fetal Therapy, Ospedale Pediatrico Microcitemico “A.Cao,”Cagliari, Italy
| | - K. Joseph Hurt
- Divisions of Maternal Fetal Medicine and Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Federica Murgia
- Department of Prenatal and Preimplantation Genetic Diagnosis and Fetal Therapy, Ospedale Pediatrico Microcitemico “A.Cao,”Cagliari, Italy
- Clinical Metabolomics Unit, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
13
|
Kudo Y, Koh I, Sugimoto J. Localization of Indoleamine 2,3-Dioxygenase-1 and Indoleamine 2,3-Dioxygenase-2 at the Human Maternal-Fetal Interface. Int J Tryptophan Res 2020; 13:1178646920984163. [PMID: 33447047 PMCID: PMC7780199 DOI: 10.1177/1178646920984163] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 12/01/2020] [Indexed: 02/03/2023] Open
Abstract
Immunohistochemical localization of indoleamine 2,3-dioxygenase-1 and indoleamine 2,3-dioxygenase-2, the first and rate-limiting enzyme in tryptophan metabolism along the kynurenine pathway, has been studied in order to better understand the physiological significance of these enzymes at the maternal-fetal interface of human pregnancy with a gestational age of 7 weeks (n = 1) and term placentas (37-40 weeks of gestation, n = 5). Indoleamine 2,3-dioxygenase-1 protein immunoreactivity was found in glandular epithelium of the decidua and the endothelium of the fetal blood vessels in the villous stroma with some additional positive cells in the villous core and in the decidua. The syncytiotrophoblast stained strongly for indoleamine 2,3-dioxygenase-2. Immunoreactivity of kynurenine, the immediate downstream product of indoleamine 2,3-dioxygenase-mediated tryptophan metabolism, showed the same localization as that of indoleamine 2,3-dioxygenase-1 and indoleamine 2,3-dioxygenase-2, suggesting these are functional enzymes. Interferon-γ added to placental villous explant culture markedly stimulated expression level of both mRNA and immunoreactivity of indoleamine 2,3-dioxygenase-1. The different cellular expression and interferon-γ sensitivity of these enzymes at the maternal-fetal interface suggests distinct physiological roles for each enzyme in normal human viviparity.
Collapse
Affiliation(s)
- Yoshiki Kudo
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Iemasa Koh
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Jun Sugimoto
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
14
|
Broekhuizen M, Klein T, Hitzerd E, de Rijke YB, Schoenmakers S, Sedlmayr P, Danser AHJ, Merkus D, Reiss IKM. l-Tryptophan-Induced Vasodilation Is Enhanced in Preeclampsia: Studies on Its Uptake and Metabolism in the Human Placenta. Hypertension 2020; 76:184-194. [PMID: 32475317 DOI: 10.1161/hypertensionaha.120.14970] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
l-tryptophan induces IDO (indoleamine 2,3-dioxygenase) 1-dependent vasodilation. IDO1 is expressed in placental endothelial cells and downregulated in preeclampsia. Hypothesizing that this may contribute to diminished placental perfusion, we studied l-tryptophan-induced vasodilation in healthy and early-onset preeclampsia placental arteries, focusing on placental kynurenine pathway alterations. Despite IDO1 downregulation, kynurenine pathway metabolite concentrations (measured with ultra-performance liquid chromatography-tandem mass spectrometry) were unaltered in preeclamptic versus healthy placentas. Most likely, this is due to enhanced l-tryptophan uptake, evidenced by increased l-tryptophan levels in preeclamptic placentas. Ex vivo perfused cotyledons from healthy and preeclamptic placentas released similar amounts of l-tryptophan and kynurenine pathway metabolites into the circulations. This release was not altered by adding l-tryptophan in the maternal circulation, suggesting that l-tryptophan metabolites act intracellularly. Maternally applied l-tryptophan did appear in the fetal circulation, confirming placental passage of this essential amino acid. After in vitro incubation of placental arteries with IDO1-upregulating cytokines interferon-γ and tumor necrosis factor-α, l-tryptophan induced vasodilation. This vasodilation was attenuated by both IDO1 and nitric oxide (NO) synthase inhibitors. Despite IDO1 downregulation, l-tryptophan-induced relaxation was enhanced in preeclamptic versus healthy placental arteries. However, cytokine stimulation additionally upregulated the LAT (l-type amino acid transporter) 1 in preeclamptic placental arteries only. Vasodilation to the lipophilic, transporter independent ethyl ester of l-tryptophan was reduced in preeclamptic versus healthy placental arteries, in agreement with reduced IDO1 expression. In conclusion, l-tryptophan induces IDO1- and NO-dependent relaxation in placental arteries, which is determined by l-tryptophan uptake rather than IDO1 expression. Increased l-tryptophan uptake might compensate for reduced IDO1 expression in preeclamptic placentas.
Collapse
Affiliation(s)
- Michelle Broekhuizen
- From the Division of Neonatology, Department of Pediatrics (M.B., E.H., P.S., I.K.M.R.).,Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (M.B., E.H., A.H.J.D.).,Division of Experimental Cardiology, Department of Cardiology (M.B., D.M.)
| | - Theo Klein
- Department of Clinical Chemistry (T.K., Y.B.d.R.)
| | - Emilie Hitzerd
- From the Division of Neonatology, Department of Pediatrics (M.B., E.H., P.S., I.K.M.R.).,Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (M.B., E.H., A.H.J.D.)
| | | | | | - Peter Sedlmayr
- From the Division of Neonatology, Department of Pediatrics (M.B., E.H., P.S., I.K.M.R.)
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (M.B., E.H., A.H.J.D.)
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology (M.B., D.M.).,Erasmus University Medical Center, Rotterdam, the Netherlands, Walter Brendel Center of Experimental Medicine (WBex), LMU Munich, Munich, Germany (D.M.)
| | - Irwin K M Reiss
- From the Division of Neonatology, Department of Pediatrics (M.B., E.H., P.S., I.K.M.R.)
| |
Collapse
|
15
|
Eroğlu H, Tolunay HE, Tonyalı NV, Orgul G, Şahin D, Yücel A. Comparison of placental elasticity in normal and intrauterine growth retardation pregnancies by ex vivo strain elastography. Arch Gynecol Obstet 2020; 302:109-115. [PMID: 32430754 DOI: 10.1007/s00404-020-05596-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/06/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To compare the placental elasticity in fetuses with or without intrauterine growth restriction (IUGR). MATERIALS AND METHODS One hundred pregnant women (50 IUGR and 50 healthy) with anteriorly located placenta were evaluated during the third trimester of pregnancy. Measurements were carried out by a machine that has a real-time elastographic ultrasonography feature. After obtaining the optimum image, three areas (subcutaneous tissue, center, and the edge of the placenta) were provided to identify the placental strain values. Then, the placental strain ratio (PSR) value was calculated automatically. Two groups compared in terms of their PSR values. RESULTS There was a significant difference in placental elasticity between the groups (P < 0.001). PSR value was 2.8 ± 1.2 in the IUGR group and 1.3 ± 0.6 in the control group. A PSR value of 1.78 had an 86% sensitivity (OR 4.3) and 80% specificity (OR 0.17) in IUGR cases. The positive predictive value was 81.1% and the negative predictive value was 85.1% for this cut-off value. CONCLUSIONS We have shown that placental strain ratio is increased during the third trimester of pregnancy in fetuses with IUGR. Increased stiffness and elasticity may be responsible for the onset of IUGR in some cases.
Collapse
Affiliation(s)
- Hasan Eroğlu
- Perinatology Department, Training and Research Hospital, Etlik Zubeyde Hanim Women's Health Care, Ankara, Turkey.
| | - Harun Egemen Tolunay
- Perinatology Department, Training and Research Hospital, Etlik Zubeyde Hanim Women's Health Care, Ankara, Turkey
| | - Nazan Vanlı Tonyalı
- Perinatology Department, Training and Research Hospital, Etlik Zubeyde Hanim Women's Health Care, Ankara, Turkey
| | - Gokcen Orgul
- Perinatology Department, Training and Research Hospital, Etlik Zubeyde Hanim Women's Health Care, Ankara, Turkey
| | - Dilek Şahin
- Perinatology Department, Training and Research Hospital, Etlik Zubeyde Hanim Women's Health Care, Ankara, Turkey
| | - Aykan Yücel
- Perinatology Department, Training and Research Hospital, Etlik Zubeyde Hanim Women's Health Care, Ankara, Turkey
| |
Collapse
|
16
|
Vaginal metabolome: towards a minimally invasive diagnosis of microbial invasion of the amniotic cavity in women with preterm labor. Sci Rep 2020; 10:5465. [PMID: 32214212 PMCID: PMC7096387 DOI: 10.1038/s41598-020-62542-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/13/2020] [Indexed: 11/25/2022] Open
Abstract
Microbial invasion of the amniotic cavity (MIAC) is only identified by amniocentesis, an invasive procedure that limits its clinical translation. Here, we aimed to evaluate whether the vaginal metabolome discriminates the presence/absence of MIAC in women with preterm labor (PTL) and intact membranes. We conducted a case-control study in women with symptoms of PTL below 34 weeks who underwent amniocentesis to discard MIAC. MIAC was defined as amniotic fluid positive for microorganisms identified by specific culture media. The cohort included 16 women with MIAC and 16 control (no MIAC). Both groups were matched for age and gestational age at admission. Vaginal fluid samples were collected shortly after amniocentesis. Metabolic profiles were analyzed by nuclear magnetic resonance (NMR) spectroscopy and compared using multivariate and univariate statistical analyses to identify significant differences between the two groups. The vaginal metabolomics profile of MIAC showed higher concentrations of hypoxanthine, proline, choline and acetylcholine and decreased concentrations of phenylalanine, glutamine, isoleucine, leucine and glycerophosphocholine. In conclusion, metabolic changes in the NMR-based vaginal metabolic profile are able to discriminate the presence/absence of MIAC in women with PTL and intact membranes. These metabolic changes might be indicative of enhanced glycolysis triggered by hypoxia conditions as a consequence of bacterial infection, thus explaining the utilization of alternative energy sources in an attempt to replenish glucose.
Collapse
|
17
|
Zhang L, Cao Z, Feng F, Xu YN, Li L, Gao H. A maternal GOT1 novel variant associated with early-onset severe preeclampsia identified by whole-exome sequencing. BMC MEDICAL GENETICS 2020; 21:49. [PMID: 32143588 PMCID: PMC7060644 DOI: 10.1186/s12881-020-0989-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 03/02/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND This study wants to know the genetic cause of preeclampsia (PE) which is a leading cause of maternal and perinatal death, but the underlying molecular mechanisms that cause PE remain poorly understood. Many single nucleotide polymorphisms have been identified by genome-wide association studies and were found to be associated with PE; however, few studies have used whole-exome sequencing (WES) to identify PE variants. METHODS Five patients with severe early-onset preeclampsia (EOPE) were recruited, and WES was performed on each patient. Sanger sequencing was used to confirm the potential causative genetic variant. RESULTS After a stringent bioinformatics analysis, a rare variant in the GOT1 gene, c.44C > G:p.P15R, was found in one patient. Bioinformatics analysis showed that the variant site is highly conserved across several species and was predicted to be a pathogenic variant according to several online mutational function prediction software packages. Further structural biology homology modeling suggested that P15R would change the electric environment of enzymatic center, and might affect the binding affinity of substrate or product. CONCLUSION We demonstrated for the first time that the variant in GOT1 may be associated with EOPE, the results of this study provide researchers and clinicians with a better understanding of the molecular mechanisms that underlie maternal severe EOPE.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Internal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Chaoyang, Beijing, 100026, China
| | - Zheng Cao
- Department of Laboratory Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Chaoyang, Beijing, 100026, China
| | - Fan Feng
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Haidian, Beijing, 100084, China
| | - Ya-Nan Xu
- Department of Internal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Chaoyang, Beijing, 100026, China
| | - Lin Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Chaoyang, Beijing, 100026, China.
| | - Hong Gao
- Department of Internal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Chaoyang, Beijing, 100026, China.
| |
Collapse
|
18
|
Abstract
Preeclampsia is a medical condition affecting 5-10% of pregnancies. It has serious effects on the health of the pregnant mother and developing fetus. While possible causes of preeclampsia are speculated, there is no consensus on its etiology. The advancement of big data and high-throughput technologies enables to study preeclampsia at the new and systematic level. In this review, we first highlight the recent progress made in the field of preeclampsia research using various omics technology platforms, including epigenetics, genome-wide association studies (GWAS), transcriptomics, proteomics and metabolomics. Next, we integrate the results in individual omic level studies, and show that despite the lack of coherent biomarkers in all omics studies, inhibin is a potential preeclamptic biomarker supported by GWAS, transcriptomics and DNA methylation evidence. Using network analysis on the biomarkers of all the literature reviewed here, we identify four striking sub-networks with clear biological functions supported by previous molecular-biology and clinical observations. In summary, omics integration approach offers the promise to understand molecular mechanisms in preeclampsia.
Collapse
|
19
|
Zhao L, Zheng X, Liu J, Zheng R, Yang R, Wang Y, Sun L. The placental transcriptome of the first-trimester placenta is affected by in vitro fertilization and embryo transfer. Reprod Biol Endocrinol 2019; 17:50. [PMID: 31262321 PMCID: PMC6604150 DOI: 10.1186/s12958-019-0494-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/17/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The placenta is a highly specialized temporary organ that is related to fetal development and pregnancy outcomes, and epidemiological data demonstrate an increased risk of placental abnormality after in vitro fertilization and embryo transfer (IVF-ET). METHODS This study examines alterations in the transcriptome profile of first-trimester placentas from IVF-ET pregnancies and analyzes the potential mechanisms that play a role in the adverse perinatal outcomes associated with IVF-ET procedures. Four human placental villi from first-trimester samples were obtained through fetal bud aspiration from patients subjected to IVF-ET due to oviductal factors. An additional four control human placental villi were derived from a group of subjects who spontaneously conceived a twin pregnancy. We analyzed their transcriptomes by microarray. Then, RT-qPCR and immunohistochemistry were utilized to analyze several dysregulated genes to validate the microarray results. Biological functions and pathways were analyzed with bioinformatics tools. RESULTS A total of 3405 differentially regulated genes were identified as significantly dysregulated (> 2-fold change; P < 0.05) in the IVF-ET placenta in the first trimester: 1910 upregulated and 1495 downregulated genes. Functional enrichment analysis of the differentially regulated genes demonstrated that the genes were involved in more than 50 biological processes and pathways that have been shown to play important roles in the first trimester in vivo. These pathways can be clustered into coagulation cascades, immune response, transmembrane signaling, metabolism, cell cycle, stress control, invasion and vascularization. Nearly the same number of up- and downregulated genes participate in the same biological processes related to placental development and maintenance. Procedures utilized in IVF-ET altered the expression of first-trimester placental genes that are critical to these biological processes and triggered a compensatory mechanism during early implantation in vivo. CONCLUSION These data provide a potential basis for further analysis of the higher frequency of adverse perinatal outcomes following IVF-ET, with the ultimate goal of developing safer IVF-ET protocols.
Collapse
Affiliation(s)
- Liang Zhao
- Department of Obstetrics and Gynecology, Beijing Jishuitan, Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, People's Republic of China
| | - Xiuli Zheng
- Department of Obstetrics and Gynecology, Beijing Jishuitan, Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, People's Republic of China
| | - Jingfang Liu
- Department of Obstetrics and Gynecology, Beijing Jishuitan, Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, People's Republic of China
| | - Rong Zheng
- Department of Obstetrics and Gynecology, Beijing Jishuitan, Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, People's Republic of China
| | - Rui Yang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49, Huayuan North Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Ying Wang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49, Huayuan North Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Lifang Sun
- Department of Obstetrics and Gynecology, Beijing Jishuitan, Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, People's Republic of China.
| |
Collapse
|
20
|
Worton SA, Greenwood SL, Wareing M, Heazell AE, Myers J. The kynurenine pathway; A new target for treating maternal features of preeclampsia? Placenta 2019; 84:44-49. [PMID: 31076094 DOI: 10.1016/j.placenta.2019.04.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/08/2019] [Accepted: 04/30/2019] [Indexed: 12/22/2022]
Abstract
In preeclampsia, vasospasm, oxidative stress, endothelial dysfunction, and immune dysregulation are key mediators of maternal disease. A new time-of-disease treatment is needed with the potential to treat these areas of pathophysiology. A review of the literature has indicated that metabolites of the kynurenine pathway have the potential to; (i) induce vasorelaxation of resistance arteries and reduce blood pressure; (ii) exert antioxidant effects and reduce the effects of poly-ADP ribose polymerase activation (iii) prevent endothelial dysfunction and promote endothelial nitric oxide production; (iv) cause T cell differentiation into tolerogenic regulatory T cells and induce apoptosis of pro-inflammatory Th1 cells. This has led to the hypothesis that increasing Kynurenine pathway activity may offer a new treatment strategy for preeclampsia.
Collapse
Affiliation(s)
- Stephanie A Worton
- Maternal and Fetal Health Research Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.
| | - Susan L Greenwood
- Maternal and Fetal Health Research Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Mark Wareing
- Maternal and Fetal Health Research Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Alexander Ep Heazell
- Maternal and Fetal Health Research Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; St. Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Jenny Myers
- Maternal and Fetal Health Research Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; St. Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
21
|
Wu Y, Zhang J, Peng S, Wang X, Luo L, Liu L, Huang Q, Tian M, Zhang X, Shen H. Multiple elements related to metabolic markers in the context of gestational diabetes mellitus in meconium. ENVIRONMENT INTERNATIONAL 2018; 121:1227-1234. [PMID: 30385065 DOI: 10.1016/j.envint.2018.10.044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/03/2018] [Accepted: 10/21/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is a typical fetus development niches dysfunction and many toxic/nutrient elements have been associated with its onset and progression. However, the classic epidemiologic approach is regarded as "black-box epidemiology" and fails to elucidate these elements' biological roles on the damaged fetus developmental microenvironment. OBJECTIVE We aimed to characterize the associations between meconium of multiple elements with GDM for illustrating their interruption effects on in-uterus microenvironment. METHODS In this case-control study (n = 137 cases; n = 197 controls), the participants were nested from a cross-sectional retrospection of 1359 recruitments in Xiamen, China. Twenty-one meconium elements were characterized using inductively coupled plasma mass spectrometry (ICP-MS) or inductively coupled plasma optical emission spectrometry (ICP-OES). For shifting the present paradigm from a black-box approach to a molecular approach, GDM-related metabolic markers were identified in our previous metabolome report. Based on the meet-in-middle strategy, the associations among the elements, metabolic markers and GDM incidence were assessed by using redundancy analysis and correlation-adjusted correlation; mediation analysis was further used to test the hypothesis that metabolic markers mediate the associations of the elements with GDM incidence. RESULTS Eight elements were related with the GDM occurrence in dose-dependent manners, which positively (Al, As, Ba, Cd, Hg, and Sn) or negatively (Ca and V) associated with GDM. Among them, As, Cd, Ba, and Ca significantly contributed to the variation of GDM-related metabolic markers. Additionally, the associations of Cd, Ba, Ca and As with GDM were mediated by the metabolic markers which majorly involved in the lipid metabolism and the Adenosine/l-Arginine/Nitric Oxide (ALANO) pathways. CONCLUSIONS The two-side mediations of meconium metabolic markers between the multiple elements and GDM occurrence indicated that maternal exposure to As, Ba, Cd, and Ca may be associated with the dysfunction of fetus development niche through disrupting lipid metabolism and ALANO pathways.
Collapse
Affiliation(s)
- Yan Wu
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jie Zhang
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, China
| | - Siyuan Peng
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, China
| | - Xiaofei Wang
- Department of Chemistry and The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| | - Lianzhong Luo
- Department of Pharmacy, Xiamen Medical College, Xiamen, China
| | - Liangpo Liu
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, China
| | - Qingyu Huang
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, China
| | - Meiping Tian
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, China
| | - Xueqin Zhang
- Xiamen Maternity and Child Health Care Hospital, Xiamen, China.
| | - Heqing Shen
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, China.
| |
Collapse
|
22
|
Hulme CH, Stevens A, Dunn W, Heazell AEP, Hollywood K, Begley P, Westwood M, Myers JE. Identification of the functional pathways altered by placental cell exposure to high glucose: lessons from the transcript and metabolite interactome. Sci Rep 2018; 8:5270. [PMID: 29588451 PMCID: PMC5869594 DOI: 10.1038/s41598-018-22535-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 02/19/2018] [Indexed: 02/06/2023] Open
Abstract
The specific consequences of hyperglycaemia on placental metabolism and function are incompletely understood but likely contribute to poor pregnancy outcomes associated with diabetes mellitus (DM). This study aimed to identify the functional biochemical pathways perturbed by placental exposure to high glucose levels through integrative analysis of the trophoblast transcriptome and metabolome. The human trophoblast cell line, BeWo, was cultured in 5 or 25 mM glucose, as a model of the placenta in DM. Transcriptomic analysis using microarrays, demonstrated 5632 differentially expressed gene transcripts (≥± 1.3 fold change (FC)) following exposure to high glucose. These genes were used to generate interactome models of transcript response using BioGRID (non-inferred network: 2500 nodes (genes) and 10541 protein-protein interactions). Ultra performance-liquid chromatography-mass spectrometry (MS) and gas chromatography-MS analysis of intracellular extracts and culture medium were used to assess the response of metabolite profiles to high glucose concentration. The interactions of altered genes and metabolites were assessed using the MetScape interactome database, resulting in an integrated model of systemic transcriptome (2969 genes) and metabolome (41 metabolites) response within placental cells exposed to high glucose. The functional pathways which demonstrated significant change in response to high glucose included fatty acid β-oxidation, phospholipid metabolism and phosphatidylinositol phosphate signalling.
Collapse
Affiliation(s)
- C H Hulme
- Maternal and Fetal Health Research Centre, Division of Developmental Biology & Medicine, School of Medical Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, M13 9WL, UK.,Maternal and Fetal Health Research Centre, Central Manchester University Hospitals NHS Foundation Trust, St Mary's Hospital, Manchester Academic Health sciences Centre, Manchester, M13 9WL, UK
| | - A Stevens
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine & Health University of Manchester, Manchester Academic Health Sciences Centre, Manchester, M13 9WL, UK
| | - W Dunn
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, M13 9WL, UK.,Centre for Endocrinology and Diabetes, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, M13 9WL, UK.,School of Biosciences, Phenome Centre Birmingham and Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, B15 2TT, UK
| | - A E P Heazell
- Maternal and Fetal Health Research Centre, Division of Developmental Biology & Medicine, School of Medical Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, M13 9WL, UK.,Maternal and Fetal Health Research Centre, Central Manchester University Hospitals NHS Foundation Trust, St Mary's Hospital, Manchester Academic Health sciences Centre, Manchester, M13 9WL, UK
| | - K Hollywood
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, M13 9WL, UK.,Centre for Endocrinology and Diabetes, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, M13 9WL, UK.,Manchester Institute of Biotechnology and School of Chemistry, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - P Begley
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, M13 9WL, UK.,Centre for Endocrinology and Diabetes, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, M13 9WL, UK
| | - M Westwood
- Maternal and Fetal Health Research Centre, Division of Developmental Biology & Medicine, School of Medical Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, M13 9WL, UK.,Maternal and Fetal Health Research Centre, Central Manchester University Hospitals NHS Foundation Trust, St Mary's Hospital, Manchester Academic Health sciences Centre, Manchester, M13 9WL, UK
| | - J E Myers
- Maternal and Fetal Health Research Centre, Division of Developmental Biology & Medicine, School of Medical Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, M13 9WL, UK. .,Maternal and Fetal Health Research Centre, Central Manchester University Hospitals NHS Foundation Trust, St Mary's Hospital, Manchester Academic Health sciences Centre, Manchester, M13 9WL, UK.
| |
Collapse
|
23
|
Abstract
This article presents an account of the research carried out so far in the use of metabolomics to find biomarkers of preterm birth (PTB) in fetal, maternal, and newborn biofluids. Metabolomic studies have employed mainly nuclear magnetic resonance spectroscopy or mass spectrometry-based methodologies to analyze, on one hand, prenatal biofluids (amniotic fluid, maternal urine/maternal blood, cervicovaginal fluid) to identify predictive biomarkers of PTB, and on the other hand, biofluids collected at or after birth (amniotic fluid, umbilical cord blood, newborn urine, and newborn blood, maternal blood, or breast milk) to assess and follow up the health status of PTB babies. Besides advancing on the biochemical knowledge of PTB metabolism mainly during the in utero period and at birth, the work carried out has also helped to identify important requirements related to experimental design and analytical protocol that need to be addressed, if translation of these biomarkers to the clinic is to be envisaged. An outlook of possible future developments for the translation of laboratory results to the clinic is presented.
Collapse
Affiliation(s)
- Ana M Gil
- 1 Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Daniela Duarte
- 1 Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
24
|
Global Metabolomics of the Placenta Reveals Distinct Metabolic Profiles between Maternal and Fetal Placental Tissues Following Delivery in Non-Labored Women. Metabolites 2018; 8:metabo8010010. [PMID: 29360753 PMCID: PMC5876000 DOI: 10.3390/metabo8010010] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 01/16/2018] [Accepted: 01/19/2018] [Indexed: 01/12/2023] Open
Abstract
We evaluated the metabolic alterations in maternal and fetal placental tissues from non-labored women undergoing cesarean section using samples collected from 5 min to 24 h following delivery. Using 1H-NMR, we identified 14 metabolites that significantly differed between maternal and fetal placental tissues (FDR-corrected p-value < 0.05), with 12 metabolites elevated in the maternal tissue, reflecting the flux of these metabolites from mother to fetus. In the maternal tissue, 4 metabolites were significantly altered at 15 min, 10 metabolites at 30 min, and 16 metabolites at 1 h postdelivery, while 11 metabolites remained stable over 24 h. In contrast, in the fetal placenta tissue, 1 metabolite was significantly altered at 15 min, 2 metabolites at 30 min, and 4 metabolites at 1 h postdelivery, while 22 metabolites remained stable over 24 h. Our study provides information on the metabolic profiles of maternal and fetal placental tissues delivered by cesarean section and reveals that there are different metabolic alterations in the maternal and fetal tissues of the placenta following delivery.
Collapse
|
25
|
Fattuoni C, Mandò C, Palmas F, Anelli GM, Novielli C, Parejo Laudicina E, Savasi VM, Barberini L, Dessì A, Pintus R, Fanos V, Noto A, Cetin I. Preliminary metabolomics analysis of placenta in maternal obesity. Placenta 2017; 61:89-95. [PMID: 29277276 DOI: 10.1016/j.placenta.2017.11.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 11/23/2017] [Accepted: 11/27/2017] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Metabolomics identifies phenotypical groups with specific metabolic profiles, being increasingly applied to several pregnancy conditions. This is the first preliminary study analyzing placental metabolomics in normal weight (NW) and obese (OB) pregnancies. METHODS Twenty NW (18.5 ≤ BMI< 25 kg/m2) and eighteen OB (BMI≥ 30 kg/m2) pregnancies were studied. Placental biopsies were collected at elective caesarean section. Metabolites extraction method was optimized for hydrophilic and lipophilic phases, then analyzed with GC-MS. Univariate and PLS-DA multivariate analysis were applied. RESULTS Univariate analysis showed increased uracil levels while multivariate PLS-DA analysis revealed lower levels of LC-PUFA derivatives in the lipophilic phase and several metabolites with significantly different levels in the hydrophilic phase of OB vs NW. DISCUSSION Placental metabolome analysis of obese pregnancies showed differences in metabolites involved in antioxidant defenses, nucleotide production, as well as lipid synthesis and energy production, supporting a shift towards higher placental metabolism. OB placentas also showed a specific fatty acids profile suggesting a disruption of LC-PUFA biomagnification. This study can lay the foundation to further metabolomic placental characterization in maternal obesity. Metabolic signatures in obese placentas may reflect changes occurring in the intrauterine metabolic environment, which may affect the development of adult diseases.
Collapse
Affiliation(s)
- Claudia Fattuoni
- Department of Chemical and Geological Sciences, University of Cagliari, Italy
| | - Chiara Mandò
- Unit of Obstetrics and Gynecology, Hospital "L. Sacco" and Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Italy
| | - Francesco Palmas
- Department of Chemical and Geological Sciences, University of Cagliari, Italy
| | - Gaia Maria Anelli
- Unit of Obstetrics and Gynecology, Hospital "L. Sacco" and Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Italy
| | - Chiara Novielli
- Unit of Obstetrics and Gynecology, Hospital "L. Sacco" and Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Italy
| | - Estefanìa Parejo Laudicina
- Centre of Excellence for Pediatric Research EURISTIKOS and Department of Pediatrics, School of Medicine, University of Granada, Granada, Spain
| | - Valeria Maria Savasi
- Unit of Obstetrics and Gynecology, Hospital "L. Sacco" and Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Italy
| | - Luigi Barberini
- Department of Medical Sciences and Public Health, University of Cagliari, Italy
| | - Angelica Dessì
- Maternal-Neonatal Department, Neonatal Intensive Care Unit, Puericulture Institute and Neonatal Section, AOUCA University Hospital of Cagliari, Italy
| | - Roberta Pintus
- Maternal-Neonatal Department, Neonatal Intensive Care Unit, Puericulture Institute and Neonatal Section, AOUCA University Hospital of Cagliari, Italy
| | - Vassilios Fanos
- Maternal-Neonatal Department, Neonatal Intensive Care Unit, Puericulture Institute and Neonatal Section, AOUCA University Hospital of Cagliari, Italy
| | - Antonio Noto
- Maternal-Neonatal Department, Neonatal Intensive Care Unit, Puericulture Institute and Neonatal Section, AOUCA University Hospital of Cagliari, Italy
| | - Irene Cetin
- Unit of Obstetrics and Gynecology, Hospital "L. Sacco" and Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Italy.
| |
Collapse
|
26
|
Brodowski L, Burlakov J, Hass S, von Kaisenberg C, von Versen-Höynck F. Impaired functional capacity of fetal endothelial cells in preeclampsia. PLoS One 2017; 12:e0178340. [PMID: 28542561 PMCID: PMC5441640 DOI: 10.1371/journal.pone.0178340] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 05/11/2017] [Indexed: 12/02/2022] Open
Abstract
Objectives Preeclampsia is one of the main contributers to maternal and fetal morbidity and mortality during pregnancy. A history of preeclampsia puts mother and offspring at an increased cardiovascular risk in later life. We hypothesized that at the time of birth functional impairments of fetal endothelial cells can be detected in pregnancies complicated by preeclampsia and that a therapeutic intervention using 1,25 (OH)2 vitamin D3 can reverse the adverse effects of preeclampsia on cell function. Methods Human umbilical vein endothelial cells (HUVEC) were isolated from umbilical cords obtained from preeclamptic (N = 12) and uncomplicated pregnancies (N = 13, control). Placental villous tissue fragments from uncomplicated term pregnancies were incubated in explant culture for 48 h at 2% (hypoxia), 8% or 21% O2. Explant conditioned media (CM) was collected and pooled according to oxygen level. We compared the ability of preeclampsia vs. control HUVEC to migrate, proliferate, and form tubule-like networks in a Matrigel assay, in the presence/absence of CM and 1,25(OH)2 vitamin D3. Results HUVEC from preeclamptic pregnancies showed reduced migration (P = 0.04) and tubule formation (P = 0.04), but no change in proliferation (P = 0.16) compared to healthy pregnancies. Placental villous explant CM derived from 2% O2 incubations significantly reduced HUVEC migration, when compared to non-CM (P = 0.04). Vitamin D3 improved HUVEC function in neither of the groups. There was no significant difference in VEGF gene expression between healthy and preeclamptic pregnancies and no effect of Vitamin D3 on VEGF expression. Conclusions Reduced functional abilities of fetal endothelial cells from preeclamptic pregnancies suggests that disease pathways, possibly originating from the dysfunctional placenta, negatively impact fetal endothelium. The neutral effect of 1,25(OH)2 vitamin D3 contrasts with previous findings that vitamin D rescues the poor migration, proliferation and tubule formation exhibited by cord blood fetal endothelial progenitor cells from preeclamptic pregnancies. Further investigations to distinguish pathways by which offspring exposed to preeclampsia are at risk for cardiovascular disease are needed.
Collapse
Affiliation(s)
- Lars Brodowski
- Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Jennifer Burlakov
- Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Sarah Hass
- Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | | | | |
Collapse
|
27
|
Song F, Wu W, Qian Z, Zhang G, Cheng Y. Assessment of the Placenta in Intrauterine Growth Restriction by Diffusion-Weighted Imaging and Proton Magnetic Resonance Spectroscopy. Reprod Sci 2017; 24:575-581. [PMID: 27647687 DOI: 10.1177/1933719116667219] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Placental insufficiency is a major cause of intrauterine growth restriction (IUGR) and seriously affects fetal development. When placental insufficiency happened, the diffusion of water molecule was restricted and the metabolic balance was destroyed in the placenta. In this prospective study, we aimed to evaluate the diagnostic value of diffusion-weighted imaging (DWI) in combination with proton magnetic resonance spectroscopy (1H MRS) for placental insufficiency in IUGR. The apparent diffusion coefficient (ADC) values were calculated using DWI, and the metabolism of N-acetylaspartate (NAA), choline, and lipid and their ratios in the placenta were calculated using 1H MRS. Data were statistically analyzed by receiver operating characteristic (ROC) curves and logistic regression analysis. The NAA and choline peaks were decreased in IUGR placentas compared with normal placentas, while lipid peaks showed an opposite trend. The average ADC value and the NAA/lipid and choline/lipid ratios were lower in the IUGR group than in the normal group ( P < .01). Logistic regression with multiple parameters showed that combination of the ADC value and choline/lipid ratio was more favorable than either parameter alone in analyzing placental insufficiency of IUGR ( P < .05). The area under the ROC curve for the combination was 0.939, indicating reasonably good discrimination. We concluded that reduced ADC and NAA/lipid and choline/lipid ratios could serve as potential markers for placental insufficiency of IUGR. Furthermore, the combination of ADC value and choline/lipid ratio greatly improved the diagnostic value.
Collapse
Affiliation(s)
- Fuzhen Song
- 1 Department of Radiology, The Tenth People's Hospital of Tong Ji University, Shanghai, China
- 2 Department of Radiology, The International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weibin Wu
- 2 Department of Radiology, The International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaoxia Qian
- 2 Department of Radiology, The International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guofu Zhang
- 3 Department of Radiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yingsheng Cheng
- 1 Department of Radiology, The Tenth People's Hospital of Tong Ji University, Shanghai, China
- 4 Department of Radiology, The Sixth Affiliated Hospital of Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
28
|
Characterizing the lipid and metabolite changes associated with placental function and pregnancy complications using ion mobility spectrometry-mass spectrometry and mass spectrometry imaging. Placenta 2017; 60 Suppl 1:S67-S72. [PMID: 28392013 DOI: 10.1016/j.placenta.2017.03.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/23/2017] [Accepted: 03/28/2017] [Indexed: 11/23/2022]
Abstract
Successful pregnancy is dependent upon discrete biological events, which include embryo implantation, decidualization, and placentation. Problems associated with each of these events can cause infertility or conditions such as preeclampsia. A greater understanding of the molecular changes associated with these complex processes is necessary to aid in identifying treatments for each condition. Previous nuclear magnetic resonance spectroscopy and mass spectrometry studies have been used to identify metabolites and lipids associated with pregnancy-related complications. However, due to limitations associated with conventional implementations of both techniques, novel technology developments are needed to more fully understand the initiation and development of pregnancy related problems at the molecular level. In this perspective, we describe current analytical techniques for metabolomic and lipidomic characterization of pregnancy complications and discuss the potential for new technologies such as ion mobility spectrometry-mass spectrometry and mass spectrometry imaging to contribute to a better understanding of the molecular changes that affect the placenta and pregnancy outcomes.
Collapse
|
29
|
Murray PG, Butcher I, Dunn WB, Stevens A, Perchard R, Hanson D, Whatmore A, Westwood M, Clayton PE. Metabolites involved in glycolysis and amino acid metabolism are altered in short children born small for gestational age. Pediatr Res 2016; 80:299-305. [PMID: 27057740 PMCID: PMC4939268 DOI: 10.1038/pr.2016.72] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 02/02/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND Later life metabolic dysfunction is a well-recognized consequence of being born small for gestational age (SGA). This study has applied metabolomics to identify whether there are changes in these pathways in prepubertal short SGA children and aimed to compare the intracellular and extracellular metabolome in fibroblasts derived from healthy children and SGA children with postnatal growth impairment. METHODS Skin fibroblast cell lines were established from eight SGA children (age 1.8-10.3 y) with failure of catch-up growth and from three healthy control children. Confluent cells were incubated in serum-free media and the spent growth medium (metabolic footprint), and intracellular metabolome (metabolic fingerprint) were analyzed by gas-chromatography mass spectrometry. RESULTS Nineteen metabolites were significantly altered between SGA and control cell lines. The greatest fold difference (FD) was seen for alanine (fingerprint FD, SGA: control 0.3, P = 0.01 and footprint FD = 0.19, P = 0.01), aspartic acid (fingerprint FD = 5.21, P = 0.01), and cystine (footprint FD = 1.66, P = 0.02). Network analysis of the differentially expressed metabolites predicted inhibition of insulin as well as growth (ERK) signaling in SGA cells. CONCLUSION This study indicates that changes in cellular metabolism associated with both growth failure and insulin insensitivity are present in prepubertal short children born SGA.
Collapse
Affiliation(s)
- Philip G Murray
- Centres for Paediatrics and Child Health, Institute of Human Development, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Oxford Road, Manchester, M13 9WL, UK.
| | - Imogen Butcher
- Centres for Paediatrics and Child Health, Institute of Human Development, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Oxford Road, Manchester, M13 9WL, UK.
| | - Warwick B Dunn
- Centre for Advanced Discovery & Experimental Therapeutics (CADET), Institute of Human Development, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Nowgen Centre, Grafton Street, Manchester, M13 9WU, UK.
,Manchester Centre for Integrative Systems Biology, School of Chemistry, University of Manchester, Princess Street, Manchester, M1 7DN, UK
,School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Adam Stevens
- Centres for Paediatrics and Child Health, Institute of Human Development, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Oxford Road, Manchester, M13 9WL, UK.
| | - Reena Perchard
- Centres for Paediatrics and Child Health, Institute of Human Development, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Oxford Road, Manchester, M13 9WL, UK.
| | - Daniel Hanson
- Centres for Paediatrics and Child Health, Institute of Human Development, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Oxford Road, Manchester, M13 9WL, UK.
| | - Andrew Whatmore
- Centres for Paediatrics and Child Health, Institute of Human Development, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Oxford Road, Manchester, M13 9WL, UK.
| | - Melissa Westwood
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, St Mary’s Hospital, Manchester, Oxford Road, Manchester, M13 9WL, UK.
| | - Peter E Clayton
- Centres for Paediatrics and Child Health, Institute of Human Development, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Oxford Road, Manchester, M13 9WL, UK.
| |
Collapse
|
30
|
Dunn WB, Allwood JW, Van Mieghem T, Morris RK, Mackie FL, Fox CE, Kilby MD. Carbohydrate and fatty acid perturbations in the amniotic fluid of the recipient twin of pregnancies complicated by twin-twin transfusion syndrome in relation to treatment and fetal cardiovascular risk. Placenta 2016; 44:6-12. [DOI: 10.1016/j.placenta.2016.05.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 05/23/2016] [Accepted: 05/26/2016] [Indexed: 10/21/2022]
|
31
|
Kedia K, Smith SF, Wright AH, Barnes JM, Tolley HD, Esplin MS, Graves SW. Global "omics" evaluation of human placental responses to preeclamptic conditions. Am J Obstet Gynecol 2016; 215:238.e1-238.e20. [PMID: 26970495 DOI: 10.1016/j.ajog.2016.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 02/29/2016] [Accepted: 03/02/2016] [Indexed: 10/22/2022]
Abstract
BACKGROUND Preeclampsia (PE) is a leading cause of maternal death. Its cause is still debated but there is general agreement that the placenta plays a central role. Perhaps the most commonly proposed contributors to PE include placental hypoxia, oxidative stress, and increased proinflammatory cytokines. How the placenta responds to these abnormalities has been considered but not as part of a comprehensive analysis of low-molecular-weight biomolecules and their responses to these accepted PE conditions. OBJECTIVE Using a peptidomic approach, we sought to identify a set of molecules exhibiting differential expression in consequence of provocative agents/chemical mediators of PE applied to healthy human placental tissue. STUDY DESIGN Known PE conditions were imposed on normal placental tissue from 13 uncomplicated pregnancies and changes in the low-molecular-weight peptidome were evaluated. A t test was used to identify potential markers for each imposed stress. These markers were then submitted to a least absolute shrinkage and selection operator multinomial logistic regression model to identify signatures specific to each stressor. Estimates of model performance on external data were obtained through internal validation. RESULTS A total of 146 markers were increased/decreased as a consequence of exposure to proposed mediators of PE. Of these 75 changed with hypoxia; 23 with hypoxia-reoxygenation/oxidative stress and 48 from exposure to tumor necrosis factor-α. These markers were chemically characterized using tandem mass spectrometry. Identification rates were: hypoxia, 34%; hypoxia-reoxygenation, 60%; and tumor necrosis factor-α, 50%. Least absolute shrinkage and selection operator modeling specified 16 markers that effectively distinguished all groups, ie, the 3 abnormal conditions and control. Bootstrap estimates of misclassification rates, multiclass area under the curve, and Brier score were 0.108, 0.944, and 0.160, respectively. CONCLUSION Using this approach we found previously unknown molecular changes in response to individual PE conditions that allowed development biomolecular signatures for exposure to each accepted pathogenic condition.
Collapse
|
32
|
Austdal M, Thomsen LCV, Tangerås LH, Skei B, Mathew S, Bjørge L, Austgulen R, Bathen TF, Iversen AC. Metabolic profiles of placenta in preeclampsia using HR-MAS MRS metabolomics. Placenta 2015; 36:1455-62. [PMID: 26582504 DOI: 10.1016/j.placenta.2015.10.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Preeclampsia is a heterogeneous gestational disease characterized by maternal hypertension and proteinuria, affecting 2-7% of pregnancies. The disorder is initiated by insufficient placental development, but studies characterizing the placental disease components are lacking. METHODS Our aim was to phenotype the preeclamptic placenta using high-resolution magic angle spinning nuclear magnetic resonance spectroscopy (HR-MAS MRS). Placental samples collected after delivery from women with preeclampsia (n = 19) and normotensive pregnancies (n = 15) were analyzed for metabolic biomarkers including amino acids, osmolytes, and components of the energy and phospholipid metabolism. The metabolic biomarkers were correlated to clinical characteristics and inflammatory biomarkers in the maternal sera. RESULTS Principal component analysis showed inherent differences in placental metabolic profiles between preeclamptic and normotensive pregnancies. Significant differences in metabolic profiles were found between placentas from severe and non-severe preeclampsia, but not between preeclamptic pregnancies with fetal growth restricted versus normal weight neonates. The placental metabolites correlated with the placental stress marker sFlt-1 and triglycerides in maternal serum, suggesting variation in placental stress signaling between different placental phenotypes. DISCUSSION HR-MAS MRS is a sensitive method for defining the placental disease component of preeclampsia, identifying several altered metabolic pathways. Placental HR-MAS MRS analysis may improve insight into processes affected in the preeclamptic placenta, and represents a novel long-required tool for a sensitive placental phenotyping of this heterogeneous disease.
Collapse
Affiliation(s)
- Marie Austdal
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway; St. Olavs Hospital, Trondheim University Hospital, 7006 Trondheim, Norway; Centre of Molecular Inflammation Research, and Department of Cancer Research and Molecular Medicine, NTNU, 7491 Trondheim, Norway.
| | - Liv Cecilie Vestrheim Thomsen
- Department of Gynecology and Obstetrics, Haukeland University Hospital, 5021 Bergen, Norway; Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; Centre of Molecular Inflammation Research, and Department of Cancer Research and Molecular Medicine, NTNU, 7491 Trondheim, Norway.
| | - Line Haugstad Tangerås
- St. Olavs Hospital, Trondheim University Hospital, 7006 Trondheim, Norway; Centre of Molecular Inflammation Research, and Department of Cancer Research and Molecular Medicine, NTNU, 7491 Trondheim, Norway.
| | - Bente Skei
- Centre of Molecular Inflammation Research, and Department of Cancer Research and Molecular Medicine, NTNU, 7491 Trondheim, Norway.
| | - Seema Mathew
- Department of Gynecology and Obstetrics, Haukeland University Hospital, 5021 Bergen, Norway.
| | - Line Bjørge
- Department of Gynecology and Obstetrics, Haukeland University Hospital, 5021 Bergen, Norway; Department of Clinical Science, University of Bergen, 5021 Bergen, Norway.
| | - Rigmor Austgulen
- Centre of Molecular Inflammation Research, and Department of Cancer Research and Molecular Medicine, NTNU, 7491 Trondheim, Norway.
| | - Tone Frost Bathen
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway.
| | - Ann-Charlotte Iversen
- Centre of Molecular Inflammation Research, and Department of Cancer Research and Molecular Medicine, NTNU, 7491 Trondheim, Norway.
| |
Collapse
|
33
|
Novel “omics” approach for study of low-abundance, low-molecular-weight components of a complex biological tissue: regional differences between chorionic and basal plates of the human placenta. Anal Bioanal Chem 2015; 407:8543-56. [DOI: 10.1007/s00216-015-9009-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 08/04/2015] [Accepted: 08/26/2015] [Indexed: 10/23/2022]
|
34
|
Women with preterm birth have a distinct cervicovaginal metabolome. Am J Obstet Gynecol 2015; 212:776.e1-776.e12. [PMID: 25827503 DOI: 10.1016/j.ajog.2015.03.052] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 03/16/2015] [Accepted: 03/26/2015] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Metabolomics has the potential to reveal novel pathways involved in the pathogenesis of preterm birth (PTB). The objective of this study was to investigate whether the cervicovaginal (CV) metabolome was different in asymptomatic women destined to have a PTB compared with term birth. STUDY DESIGN A nested case-control study was performed using CV fluid collected from a larger prospective cohort. The CV fluid was collected between 20-24 weeks (V1) and 24-28 weeks (V2). The metabolome was compared between women with a spontaneous PTB (n = 10) to women who delivered at term (n = 10). Samples were extracted and prepared for analysis using a standard extraction solvent method. Global biochemical profiles were determined using gas chromatography/mass spectrometry and ultra-performance liquid chromatography/tandem mass spectrometry. An ANOVA was used to detect differences in biochemical compounds between the groups. A false discovery rate was estimated to account for multiple comparisons. RESULTS A total of 313 biochemicals were identified in CV fluid. Eighty-two biochemicals were different in the CV fluid at V1 in those destined to have a PTB compared with term birth, whereas 48 were different at V2. Amino acid, carbohydrate, and peptide metabolites were distinct between women with and without PTB. CONCLUSION These data suggest that the CV space is metabolically active during pregnancy. Changes in the CV metabolome may be observed weeks, if not months, prior to any clinical symptoms. Understanding the CV metabolome may hold promise for unraveling the pathogenesis of PTB and may provide novel biomarkers to identify women most at risk.
Collapse
|
35
|
Pantham P, Heazell AE, Mullard G, Begley P, Chen Q, Brown M, Dunn WB, Chamley LW. Antiphospholipid Antibodies Alter Cell-Death-Regulating Lipid Metabolites in First and Third Trimester Human Placentae. Am J Reprod Immunol 2015; 74:181-99. [DOI: 10.1111/aji.12387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 03/13/2015] [Indexed: 02/06/2023] Open
Affiliation(s)
- Priyadarshini Pantham
- Department of Obstetrics & Gynaecology; The University of Auckland; Auckland New Zealand
- Section of Neonatology; Department of Pediatrics; University of Colorado Anschutz Medical Campus; Aurora CO USA
| | - Alexander E.P. Heazell
- The Maternal and Fetal Health Research Centre; St. Mary's Hospital; The University of Manchester; Manchester UK
| | - Graham Mullard
- Centre for Advanced Discovery and Experimental Therapeutics; Manchester Biomedical Research Centre; The University of Manchester; Manchester UK
| | - Paul Begley
- Centre for Advanced Discovery and Experimental Therapeutics; Manchester Biomedical Research Centre; The University of Manchester; Manchester UK
| | - Qi Chen
- Department of Obstetrics & Gynaecology; The University of Auckland; Auckland New Zealand
| | - Maria Brown
- Centre for Advanced Discovery and Experimental Therapeutics; Manchester Biomedical Research Centre; The University of Manchester; Manchester UK
| | - Warwick B. Dunn
- Centre for Advanced Discovery and Experimental Therapeutics; Manchester Biomedical Research Centre; The University of Manchester; Manchester UK
- Centre for Endocrinology and Diabetes; Institute of Human Development; The University of Manchester; Manchester UK
- School of Biosciences; The University of Birmingham; Edgbaston Birmingham UK
| | - Lawrence W. Chamley
- Department of Obstetrics & Gynaecology; The University of Auckland; Auckland New Zealand
| |
Collapse
|
36
|
Jadoon A, Cunningham P, McDermott LC. Regulation of fatty acid binding proteins by hypoxia inducible factors 1α and 2α in the placenta: relevance to pre-eclampsia. Prostaglandins Leukot Essent Fatty Acids 2015; 93:25-9. [PMID: 25305177 DOI: 10.1016/j.plefa.2014.09.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 08/23/2014] [Accepted: 09/16/2014] [Indexed: 10/24/2022]
Abstract
Pre-eclampsia is characterized by placental hypoxia and dyslipidemia. Arachidonic and docosahexanoic acids are essential maternal nutrients for fetal development. They are transported via placental trophoblast cells by membrane and cytosolic fatty acid binding proteins. Others report the expressions of these proteins which are increased in hypoxic trophoblasts. Using bioinformatics, BeWo cells, reporter assays, quantitative real-time PCR and immunoblotting we tested the hypothesis that hypoxia inducible factors 1α (HIF-1α) and/or 2α (HIF-2α) regulate the expressions of FABP1, FABP3, FABP4 and FATP2 proteins. Three hypoxia responsive elements (HRE) were identified in FABP1 which cumulatively responded strongly to HIF-1α and weakly to HIF-2α. FABP3 expression partially responded to HIF-1α. Two putative HRE were validated in FABP4 both of which responded weakly to HIF-1α and HIF-2α. FATP2 protein expression reacted positively to hypoxia. Thus, fetal essential fatty acid supply via the placenta is protected under hypoxia. It will be interesting to determine if our findings are replicated in human pre-eclamptic placenta.
Collapse
Affiliation(s)
- Ayesha Jadoon
- Diabetes and Nutritional Sciences Division, School of Medicine, United Kingdom
| | - Phil Cunningham
- Department of Biochemistry, School of Biomedical and Health Sciences, King׳s College London, Franklin Wilkins Building, Stamford Street, London SE1 9NH, United Kingdom
| | - Lindsay C McDermott
- Diabetes and Nutritional Sciences Division, School of Medicine, United Kingdom.
| |
Collapse
|
37
|
Metabolomics in the developmental origins of obesity and its cardiometabolic consequences. J Dev Orig Health Dis 2015; 6:65-78. [PMID: 25631626 DOI: 10.1017/s204017441500001x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In this review, we discuss the potential role of metabolomics to enhance understanding of obesity-related developmental origins of health and disease (DOHaD). We first provide an overview of common techniques and analytical approaches to help interested investigators dive into this relatively novel field. Next, we describe how metabolomics may capture exposures that are notoriously difficult to quantify, and help to further refine phenotypes associated with excess adiposity and related metabolic sequelae over the life course. Together, these data can ultimately help to elucidate mechanisms that underlie fetal metabolic programming. Finally, we review current gaps in knowledge and identify areas where the field of metabolomics is likely to provide insights into mechanisms linked to DOHaD in human populations.
Collapse
|
38
|
Metabolomics and the great obstetrical syndromes--GDM, PET, and IUGR. Best Pract Res Clin Obstet Gynaecol 2014; 29:156-64. [PMID: 25271062 DOI: 10.1016/j.bpobgyn.2014.04.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 04/13/2014] [Indexed: 01/05/2023]
Abstract
Gestational diabetes mellitus, intrauterine growth restriction, and preeclamptic toxemia are common pregnancy complications that can have detrimental effects on morbidity and mortality of the mother and fetus as well as long-term health outcomes. Although they are distinct conditions, they may occur together and are often considered together as they share a common etiology of inadequate placental perfusion. The discovery and study of preventative treatments is hampered by a lack of effective screening tools to accurately identify women at the highest risk of disease. Metabolomics, an omic science, is the global quantitative assessment of endogenous metabolites within a biological system. It has proven to be a rapid approach in the identification of biomarkers predictive of the outcome of a pathological condition and the individual's response to a pharmacological treatment. We review the current and potential applications of metabolomics in maternal-fetal medicine, focusing on its use as a biomarker for great obstetrical syndromes diagnosis.
Collapse
|
39
|
Brodowski L, Burlakov J, Myerski AC, von Kaisenberg CS, Grundmann M, Hubel CA, von Versen-Höynck F. Vitamin D prevents endothelial progenitor cell dysfunction induced by sera from women with preeclampsia or conditioned media from hypoxic placenta. PLoS One 2014; 9:e98527. [PMID: 24887145 PMCID: PMC4041729 DOI: 10.1371/journal.pone.0098527] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 05/01/2014] [Indexed: 01/17/2023] Open
Abstract
CONTEXT Placenta-derived circulating factors contribute to the maternal endothelial dysfunction underlying preeclampsia. Endothelial colony forming cells (ECFC), a sub-population of endothelial progenitor cells (EPCs), are thought to be involved in vasculogenesis and endothelial repair. Low vitamin D concentrations are associated with an increased risk for preeclampsia. OBJECTIVE We hypothesized that the function of human fetal ECFCs in culture would be suppressed by exposure to preeclampsia-related factors--preeclampsia serum or hypoxic placental conditioned medium--in a fashion reversed by vitamin D. DESIGN, SETTING, PATIENTS ECFCs were isolated from cord blood of uncomplicated pregnancies and expanded in culture. Uncomplicated pregnancy villous placenta in explant culture were exposed to either 2% (hypoxic), 8% (normoxic) or 21% (hyperoxic) O2 for 48 h, after which the conditioned media (CM) was collected. OUTCOME MEASURES ECFC tubule formation (Matrigel assay) and migration were examined in the presence of either maternal serum from preeclampsia cases or uncomplicated pregnancy controls, or pooled CM, in the presence or absence of 1,25(OH)2 vitamin D3. RESULTS 1,25(OH)2 vitamin D3 reversed the adverse effects of preeclampsia serum or CM from hypoxic placenta on ECFCs capillary-tube formation and migration. Silencing of VDR expression by VDR siRNA, VDR blockade, or VEGF pathway blockade reduced ECFC functional abilities. Effects of VDR or VEGF blockade were partially prevented by vitamin D. CONCLUSION Vitamin D promotes the capillary-like tubule formation and migration of ECFCs in culture, minimizing the negative effects of exposure to preeclampsia-related factors. Further evaluation of the role of vitamin D in ECFC regulation and preeclampsia is warranted.
Collapse
Affiliation(s)
- Lars Brodowski
- Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Jennifer Burlakov
- Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Ashley C. Myerski
- Magee- Womens Research Institute and Foundation, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | | | - Magdalena Grundmann
- Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Carl A. Hubel
- Magee- Womens Research Institute and Foundation, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | | |
Collapse
|
40
|
Mathiesen L, Mørck TA, Zuri G, Andersen MH, Pehrson C, Frederiksen M, Mose T, Rytting E, Poulsen MS, Nielsen JKS, Knudsen LE. Modelling of human transplacental transport as performed in Copenhagen, Denmark. Basic Clin Pharmacol Toxicol 2014; 115:93-100. [PMID: 24646015 DOI: 10.1111/bcpt.12228] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/04/2014] [Indexed: 12/21/2022]
Abstract
Placenta perfusion models are very effective when studying the placental mechanisms in order to extrapolate to real-life situations. The models are most often used to investigate the transport of substances between mother and foetus, including the potential metabolism of these. We have studied the relationships between maternal and foetal exposures to various compounds including pollutants such as polychlorinated biphenyls, polybrominated flame retardants, nanoparticles as well as recombinant human antibodies. The compounds have been studied in the human placenta perfusion model and to some extent in vitro with an established human monolayer trophoblast cell culture model. Results from our studies distinguish placental transport of substances by physicochemical properties, adsorption to placental tissue, binding to transport and receptor proteins and metabolism. We have collected data from different classes of chemicals and nanoparticles for comparisons across chemical structures as well as different test systems. Our test systems are based on human material to bypass the extrapolation from animal data. By combining data from our two test systems, we are able to rank and compare the transport of different classes of substances according to their transport ability. Ultimately, human data including measurements in cord blood contribute to the study of placental transport.
Collapse
Affiliation(s)
- Line Mathiesen
- Section of Environmental Health, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Khaustova SA, Senyavina NV, Tonevitsky AG, Eremina OV, Pavlovich SV. Serum metabolic profiles of pregnant women with burdened obstetrical history. Bull Exp Biol Med 2013; 156:98-100. [PMID: 24319740 DOI: 10.1007/s10517-013-2287-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The content of low-molecular-weight components in blood serum was studied by tandem mass-spectrometry in pregnant women. Serum metabolic profiles of patients with a grave obstetrical history were detected. The most significant changes were observed for the concentrations of low-molecular-weight substances involved in glucogenesis and β-oxidation processes and in metabolic chains involving carbohydrates, carnitines, amino acids, and lipids.
Collapse
Affiliation(s)
- S A Khaustova
- BioClinicum Research and Development Center; Research Institute of Pathology and Pathophysiology, the Russian Academy of Medical Sciences; V. I. Kulakov Research Center of Obstetrics, Gynecology, and Perinatology, the Ministry of Health and Social Development of the Russian Federation, Moscow, Russia.
| | | | | | | | | |
Collapse
|
42
|
AlRabiah H, Correa E, Upton M, Goodacre R. High-throughput phenotyping of uropathogenic E. coli isolates with Fourier transform infrared spectroscopy. Analyst 2013; 138:1363-9. [PMID: 23325321 DOI: 10.1039/c3an36517d] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Fourier transform infrared (FT-IR) spectroscopy is an established rapid whole-organism fingerprinting method that generates metabolic fingerprints from bacteria that reflect the phenotype of the microorganism under investigation. However, whilst FT-IR spectroscopy is fast (typically 10 s to 1 min per sample), the approaches for microbial sample preparation can be time consuming as plate culture or shake flasks are used for growth of the organism. We report a new approach that allows micro-cultivation of bacteria from low volumes (typically 200 μL) to be coupled with FT-IR spectroscopy. This approach is fast and easy to perform and gives equivalent data to the lengthier and more expensive shake flask cultivations (sample volume = 20 mL). With this micro-culture approach we also demonstrate high reproducibility of the metabolic fingerprints. The approach allowed separation of different isolates of Escherichia coli involved in urinary tract infection, including members of the globally disseminated ST131 clone, with respect to both genotype and resistance or otherwise to the antibiotic Ciprofloxacin.
Collapse
Affiliation(s)
- Haitham AlRabiah
- School of Chemistry and Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | | | | | | |
Collapse
|
43
|
Liu C, Wang W, Parchim N, Irani RA, Blackwell SC, Sibai B, Jin J, Kellems RE, Xia Y. Tissue transglutaminase contributes to the pathogenesis of preeclampsia and stabilizes placental angiotensin receptor type 1 by ubiquitination-preventing isopeptide modification. Hypertension 2013; 63:353-61. [PMID: 24191290 DOI: 10.1161/hypertensionaha.113.02361] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Preeclampsia is a life-threatening pregnancy disorder that is widely thought to be triggered by impaired placental development. However, the placenta-related pathogenic factors are not fully identified, and their underlying mechanisms in disease development remain unclear. Here, we report that the protein level and enzyme activity of tissue transglutaminase (TG2 or tTG), the most ubiquitous member of a family of enzymes that conducts post-translational modification of proteins by forming ε-(γ-glutamyl)-lysine isopeptide bonds, are significantly elevated in placentas of preeclamptic women. TG2 is localized in the placental syncytiotrophoblasts of patients with preeclampsia where it catalyzes the isopeptide modification of the angiotensin receptor type 1 (AT1). To determine the role of elevated TG2 in preeclampsia, we used a mouse model of preeclampsia based on injection of AT1-agonistic autoantibody. A pathogenic role for TG2 in preeclampsia is suggested by in vivo experiments in which cystamine, a potent transglutaminase inhibitor, or small interfering RNA-mediated TG2 knockdown significantly attenuated autoantibody-induced hypertension and proteinuria in pregnant mice. Cystamine treatment also prevented isopeptide modification of placental AT1 receptors in preeclamptic mice. Mechanistically, we revealed that AT1-agonistic autoantibody stimulation enhances the interaction between AT1 receptor and TG2 and results in increased AT1 receptor stabilization via transglutaminase-mediated isopeptide modification in trophoblasts. Mutagenesis studies further demonstrated that TG2-mediated isopeptide modification of AT1 receptors prevents ubiquitination-dependent receptor degradation. Taken together, our studies not only identify a novel pathogenic involvement of TG2 in preeclampsia but also suggest a previously unrecognized role of TG2 in the regulation of G protein-coupled receptor stabilization by inhibiting ubiquitination-dependent degradation.
Collapse
Affiliation(s)
- Chen Liu
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas. or
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Metabolomics application in maternal-fetal medicine. BIOMED RESEARCH INTERNATIONAL 2013; 2013:720514. [PMID: 23841090 PMCID: PMC3690726 DOI: 10.1155/2013/720514] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 05/10/2013] [Accepted: 05/13/2013] [Indexed: 12/24/2022]
Abstract
Metabolomics in maternal-fetal medicine is still an “embryonic” science. However, there is already an increasing interest in metabolome of normal and complicated pregnancies, and neonatal outcomes. Tissues used for metabolomics interrogations of pregnant women, fetuses and newborns are amniotic fluid, blood, plasma, cord blood, placenta, urine, and vaginal secretions. All published papers highlight the strong correlation between biomarkers found in these tissues and fetal malformations, preterm delivery, premature rupture of membranes, gestational diabetes mellitus, preeclampsia, neonatal asphyxia, and hypoxic-ischemic encephalopathy. The aim of this review is to summarize and comment on original data available in relevant published works in order to emphasize the clinical potential of metabolomics in obstetrics in the immediate future.
Collapse
|
45
|
Alexandre-Gouabau MC, Courant F, Moyon T, Küster A, Le Gall G, Tea I, Antignac JP, Darmaun D. Maternal and cord blood LC-HRMS metabolomics reveal alterations in energy and polyamine metabolism, and oxidative stress in very-low birth weight infants. J Proteome Res 2013; 12:2764-78. [PMID: 23527880 DOI: 10.1021/pr400122v] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
To assess the global effect of preterm birth on fetal metabolism and maternal-fetal nutrient transfer, we used a mass spectrometric-based chemical phenotyping approach on cord blood obtained at the time of birth. We sampled umbilical venous, umbilical arterial, and maternal blood from mothers delivering very-low birth weight (VLBW, with a median gestational age and weight of 29 weeks, and 1210 g, respectively) premature or full-term (FT) neonates. In VLBW group, we observed a significant elevation in the levels and maternal-fetal gradients of butyryl-, isovaleryl-, hexanoyl- and octanoyl-carnitines, suggesting enhanced short- and medium chain fatty acid β-oxidation in human preterm feto-placental unit. The significant decrease in glutamine-glutamate in preterm arterial cord blood beside lower levels of amino acid precursors of Krebs cycle suggest increased glutamine utilization in the fast growing tissues of preterm fetus with a deregulation in placental glutamate-glutamine shuttling. Enhanced glutathione utilization is likely to account for the decrease in precursor amino acids (serine, betaine, glutamate and methionine) in arterial cord blood. An increase in both the circulating levels and maternal-fetal gradients of several polyamines in their acetylated form (diacetylspermine and acetylputrescine) suggests an enhanced polyamine metabolic cycling in extreme prematurity. Our metabolomics study allowed the identification of alterations in fetal energy, antioxidant defense, and polyamines and purines flux as a signature of premature birth.
Collapse
|
46
|
Diaz SO, Barros AS, Goodfellow BJ, Duarte IF, Carreira IM, Galhano E, Pita C, Almeida MDC, Gil AM. Following Healthy Pregnancy by Nuclear Magnetic Resonance (NMR) Metabolic Profiling of Human Urine. J Proteome Res 2012; 12:969-79. [DOI: 10.1021/pr301022e] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Sílvia O. Diaz
- CICECO−Department
of Chemistry, Campus Universitário de Santiago, Universidade de Aveiro, 3810-193 Aveiro, Portugal
| | - António S. Barros
- QOPNA−Department
of Chemistry, Campus Universitário de Santiago, Universidade de Aveiro, 3810-193 Aveiro, Portugal
| | - Brian J. Goodfellow
- CICECO−Department
of Chemistry, Campus Universitário de Santiago, Universidade de Aveiro, 3810-193 Aveiro, Portugal
| | - Iola F. Duarte
- CICECO−Department
of Chemistry, Campus Universitário de Santiago, Universidade de Aveiro, 3810-193 Aveiro, Portugal
| | - Isabel M. Carreira
- Cytogenetics and Genomics Laboratory,
Faculty of Medicine, Universidade de Coimbra, Portugal and CENCIFOR - Forensic Science Centre, Portugal
- CIMAGO−Centro de Investigação Meio Ambiente, Genética e Oncobiologia, 3000 Coimbra, Portugal
| | - Eulália Galhano
- Maternity Bissaya Barreto, Centro Hospitalar de Coimbra, 3000-061 Coimbra, Portugal
| | - Cristina Pita
- Maternity Bissaya Barreto, Centro Hospitalar de Coimbra, 3000-061 Coimbra, Portugal
| | - Maria do Céu Almeida
- Maternity Bissaya Barreto, Centro Hospitalar de Coimbra, 3000-061 Coimbra, Portugal
| | - Ana M. Gil
- CICECO−Department
of Chemistry, Campus Universitário de Santiago, Universidade de Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
47
|
Denison FC, Semple SI, Stock SJ, Walker J, Marshall I, Norman JE. Novel use of proton magnetic resonance spectroscopy (1HMRS) to non-invasively assess placental metabolism. PLoS One 2012; 7:e42926. [PMID: 22900066 PMCID: PMC3416751 DOI: 10.1371/journal.pone.0042926] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 07/16/2012] [Indexed: 12/26/2022] Open
Abstract
Background Placental insufficiency is a major cause of antepartum stillbirth and fetal growth restriction (FGR). In affected pregnancies, delivery is expedited when the risks of ongoing pregnancy outweigh those of prematurity. Current tests are unable to assess placental function and determine optimal timing for delivery. An accurate, non-invasive test that clearly defines the failing placenta would address a major unmet clinical need. Proton magnetic resonance spectroscopy (1H MRS) can be used to assess the metabolic profile of tissue in-vivo. In FGR pregnancies, a reduction in N-acetylaspartate (NAA)/choline ratio and detection of lactate methyl are emerging as biomarkers of impaired neuronal metabolism and fetal hypoxia, respectively. However, fetal brain hypoxia is a late and sometimes fatal event in placental compromise, limiting clinical utility of brain 1H MRS to prevent stillbirth. We hypothesised that abnormal placental 1H MRS may be an earlier biomarker of intrauterine hypoxia, affording the opportunity to optimise timing of delivery in at-risk fetuses. Methods and Findings We recruited three women with severe placental insufficiency/FGR and three matched controls. Using a 3T MR system and a combination of phased-array coils, a 20×20×40 mm1H MRS voxel was selected along the ‘long-axis’ of the placenta with saturation bands placed around the voxel to prevent contaminant signals. A significant choline peak (choline/lipid ratio 1.35–1.79) was detected in all healthy placentae. In contrast, in pregnancies complicated by FGR, the choline/lipid ratio was ≤0.02 in all placentae, despite preservation of the lipid peak (p<0.001). Conclusions This novel proof-of-concept study suggests that in severe placental insufficiency/FGR, the observed 60-fold reduction in the choline/lipid ratio by 1H MRS may represent an early biomarker of critical placental insufficiency. Further studies will determine performance of this test and the potential role of 1H-MRS in the in-vivo assessment of placental function to inform timing of delivery.
Collapse
Affiliation(s)
- Fiona C Denison
- MRC Centre for Reproductive Health, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, Lothian, United Kingdom.
| | | | | | | | | | | |
Collapse
|
48
|
McNamara LE, Sjöström T, Meek RMD, Oreffo ROC, Su B, Dalby MJ, Burgess KEV. Metabolomics: a valuable tool for stem cell monitoring in regenerative medicine. J R Soc Interface 2012; 9:1713-24. [PMID: 22628210 PMCID: PMC3385772 DOI: 10.1098/rsif.2012.0169] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 05/01/2012] [Indexed: 02/06/2023] Open
Abstract
Metabolomics is a method for investigation of changes in the global metabolite profile of cells. This paper discusses the technical application of the approach, considering metabolite extraction, separation, mass spectrometry and data interpretation. A particular focus is on the application of metabolomics to the study of stem cell physiology in the context of biomaterials and regenerative medicine. Case studies are used to illustrate key points, focusing on the use of metabolomics in the examination of mesenchymal stem cell responses to titania-nanopillared substrata designed for orthopaedic applications.
Collapse
Affiliation(s)
- Laura E McNamara
- Centre for Cell Engineering, Division of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| | | | | | | | | | | | | |
Collapse
|
49
|
Salvolini E, Vignini A, Nanetti L, Raffaelli F, Di Primio R, Mazzanti L, Tranquilli AL. Glutamate in vitro effects on human term placental mitochondria. J Matern Fetal Neonatal Med 2012; 25:952-956. [PMID: 21740324 DOI: 10.3109/14767058.2011.601363] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Oxidative stress may affect the functionality of placental mitochondria, thus contributing to serious complications. For this reason research of protective substances is of great importance. Our aim was to evaluate, in mitochondria isolated from human term placentas, the effect of in vitro glutamate supplementation on their susceptibility to oxidation, on the chemico-physical characteristics of mitochondrial membranes, and on peroxidase and nitric oxide synthase (NOS) activities. METHODS The study was performed on mitochondria isolated from 20 healthy human term placentas. Specific exclusion criteria were: conception by assisted reproduction, chromosomal or other fetal, uterine or placental anomalies, gestational diabetes, preeclampsia, intrauterine growth restriction (IUGR), a history of smoking and hypertension, proteinuria, renal, cardiovascular, hepatic, and endocrine disease, metabolic disorders, and current infection or history of all types of infection. RESULTS Incubation with glutamate determined a reduced susceptibility to oxidative stress, an increase in mitochondrial membrane fluidity, and a decrease of both peroxidase and NOS activities. CONCLUSIONS On the basis of the observed results, we can hypothesize a role for glutamate in the control of lipid peroxidation extent in physiological pregnancies, as well as in the prevention of free radical-linked complications that can affect the health of both mother and fetus.
Collapse
Affiliation(s)
- Eleonora Salvolini
- Department of Molecular Pathology and Innovative Therapies - Histology, Polytechnic University of Marche, Ancona, Italy.
| | | | | | | | | | | | | |
Collapse
|
50
|
Heazell AEP, Bernatavicius G, Warrander L, Brown MC, Dunn WB. A metabolomic approach identifies differences in maternal serum in third trimester pregnancies that end in poor perinatal outcome. Reprod Sci 2012; 19:863-75. [PMID: 22534329 DOI: 10.1177/1933719112438446] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Metabolomics offers a powerful holistic approach to examine the metabolite composition of biofluids to identify disruptions present in disease. We used ultra performance liquid chromatography-mass spectroscopy on the maternal serum obtained in the third trimester to address the hypothesis that pregnancies ending in poor outcomes (small for gestational age infant, preterm birth, or neonatal intensive care admission, n = 40) would have a different maternal serum metabolic profiles to matched healthy pregnancies (n = 40). Ninety-eight identified metabolic features differed between normal and poor pregnancy outcomes. Classes of metabolites perturbed included free fatty acids, glycerolipids, progesterone metabolites, sterol lipids, vitamin D metabolites, and sphingolipids; these highlight potential molecular mechanisms associated with pregnancy complications in the third trimester linked by placental dysfunction. In this clinical setting, metabolomics has the potential to describe differences in fetoplacental and maternal metabolites in pregnancies with poor pregnancy outcomes compared with controls.
Collapse
Affiliation(s)
- Alexander E P Heazell
- Maternal and Fetal Health Research Centre, School of Biomedicine, University of Manchester, Manchester Academic Health Science Centre, UK.
| | | | | | | | | |
Collapse
|