1
|
Yang Q, Dai Y, Huang S, Liu B, Sun H, Zhou Y, Gong Y, Zhu F. MMEASE: enhanced analytical workflow for single-cell metabolomics. Nucleic Acids Res 2025:gkaf363. [PMID: 40308213 DOI: 10.1093/nar/gkaf363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/08/2025] [Accepted: 04/29/2025] [Indexed: 05/02/2025] Open
Abstract
Metabolomics is essential for providing an overview of what chemical processes are taking place. A clear shift from bulk metabolomics to single-cell metabolomics (SCM) is observed in current research, and an integral workflow enabling the analysis of SCM data is therefore in great demand. However, no such workflow has been available to date. Herein, MMEASE, previously designed for analyzing bulk metabolomic data, was therefore updated to its 2.0 version by developing the first comprehensive and in-depth workflow analyzing SCM data. First, it provided all sequential steps of modern SCM research (from SCM data processing, to cellular heterogeneity analysis, then to high-resolution metabolite annotation, and finally to cell-based biological interpretation). Second, compared with the existing tools, MMEASE 2.0 was superior by incorporating the widest variety of methods at every step of the SCM analyses. The originality and functionality of our MMEASE were extensively validated and explicitly described by case studies on benchmark data. All in all, MMEASE 2.0 was unique in accomplishing comprehensive and in-depth analyses of SCM data, which could be considered as an indispensable complement to the existing tools. Now, the latest version of MMEASE is freely accessible by all users at: https://idrblab.org/mmease/.
Collapse
Affiliation(s)
- Qingxia Yang
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yangbo Dai
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Shijie Huang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bing Liu
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Huaicheng Sun
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuan Zhou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yaguo Gong
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Macao 999078, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
2
|
Han XQ, Jiang HH, Chen ML, Han DY, Zhou SF, Wang JW, Ji SS, Wang LY, Lou JW, Li MQ. Gut microbiota interacting with vitamin D but not anandamide might contribute to the pathogenesis of preeclampsia: a preliminary study. Front Cell Infect Microbiol 2025; 14:1469054. [PMID: 39973918 PMCID: PMC11835824 DOI: 10.3389/fcimb.2024.1469054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/25/2024] [Indexed: 02/21/2025] Open
Abstract
Introduction Preeclampsia (PE) is a pregnancy-specific multisystem disorder and a leading cause of maternal and perinatal mortality globally. Despite numerous studies highlighting the potential roles of gut microbiota, anandamide (AEA), and Vitamin D (VitD) in PE, none have established them as reliable biomarkers for predicting disease onset. Moreover, their interactions in late-stage pregnancy women remain poorly understood. Methods Thirty-four preeclamptic patients (called PE group) and thirty-nine matched healthy late-pregnant women (called LP group) were involved in this case-control study. Fecal samples, which were used to acquire the diversity and composition of gut microbiota, were analyzed by 16S rRNA gene sequencing. Plasma AEA concentrations and serum VitD levels were determined by high-performance liquid chromatography-mass spectrometry (HPLC-MS) and liquid chromatography-tandem mass spectrometry (LC-MS/MS), respectively. Results In this study, β diversity but not α diversity significantly differed between the LP and PE groups. Compared with the LP group, the relative abundances of Prevotella, Erysipelotrichaceae_UCG-003, and Dorea were increased dramatically in the PE group, whereas the relative abundances of Subdoligranulum, Parabacteroides, Bacteroides were significantly decreased in the PE group. Furthermore, women with PE had a substantially lower plasma level of AEA and a marked decrease in serum VitD compared to normal late-pregnant women. Lastly, although the serum level of AEA was not significantly correlated with VitD or any of the top 6 marker genera, VitD was significantly negatively correlated with the relative abundance of Dorea, a novel finding in this context. Discussion The gut microbiota profile of the PE group was significantly different from that of the LP group. Although no significant correlations were identified between the plasma AEA levels and serum VitD levels or any of the top 6 identified marker genera, a significant negative correlation was observed between VitD and Dorea, indicating VitD and gut microbiota have the potential to be combined targets for early diagnosis and management of PE.
Collapse
Affiliation(s)
- Xiao-Qiang Han
- Department of Obstetrics and Gynaecology, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
- Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Xiangyang, China
| | - Hui-Hui Jiang
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai, China
| | - Meng-Ling Chen
- Department of Obstetrics and Gynaecology, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
- Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Xiangyang, China
| | - De-Yang Han
- Department of Obstetrics and Gynaecology, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
- Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Xiangyang, China
| | - Su-Fen Zhou
- Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Xiangyang, China
- Department of Ultrasound, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Jin-Wen Wang
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai, China
| | - Shu-Shen Ji
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai, China
| | - Ling-Yun Wang
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai, China
| | - Jing-Wei Lou
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai, China
| | - Ming-Qun Li
- Department of Obstetrics and Gynaecology, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
- Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Xiangyang, China
| |
Collapse
|
3
|
Zhao HJ, Chen Y, Liu T, McArthur K, Mueller NT. Short-Chain Fatty Acids and Preeclampsia: A Scoping Review. Nutr Rev 2025; 83:e683-e693. [PMID: 38796843 PMCID: PMC11723139 DOI: 10.1093/nutrit/nuae057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Preeclampsia (PE) is a pregnancy-associated hypertension disorder with high morbidity and mortality. Short-chain fatty acids (SCFAs)-molecules produced by gut microbes-have been associated with hypertension, yet their relation to PE remains uncertain. OBJECTIVES The aim was to review existing human studies that examined associations of the major SCFAs (acetate, propionate, butyrate) in pregnancy with PE development. METHODS Two reviewers independently searched online databases (EMBASE, PubMed, Web of Science, and Cochrane Database of Systematic Reviews) in January 2024 using the following terms: "short-chain fatty acids," "acetic acid," "butyric acid," "propionic acid," and "preeclampsia." The final set of included studies had to report associations of SCFAs with PE, be peer-reviewed, be written in English, and be conducted in humans. RESULTS The abstracts of 907 studies were screened; 43 underwent full-text screening and 11 (1318 total participants, 352 with PE) were included in the final review. All studies used a case-control design. SCFAs were measured in a range of biospecimens (eg, serum, plasma, feces, placentas, and amniotic fluid) that were collected at distinct time points in pregnancy. All 7 studies that investigated butyrate found that it was lower in PE cases than in controls, with 6 of these showing statistical significance (P < .05). Five studies showed that acetate was significantly lower in individuals with PE compared with healthy individuals, while 1 study found that acetate was significantly higher in PE cases. One study reported significantly higher propionate among PE cases vs controls, while 2 studies reported significantly lower propionate levels in PE cases. The nuance in results for acetate and propionate may owe to reasons such as differences in distributions of population characteristics associated with SCFA level and PE or type of PE (early vs late). CONCLUSION Current epidemiologic evidence, which derives only from case-control studies, suggests that SCFAs, particularly butyrate (protective), in pregnancy are related to the development of PE. Large-cohort studies are warranted to investigate the temporality and potential causality of these associations.
Collapse
Affiliation(s)
- Heather J Zhao
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, United States
- Temerty School of Medicine, University of Toronto, Toronto, ON M5S1A8, Canada
| | - Yingan Chen
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO 80045, United States
| | - Tiange Liu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, United States
| | - Kristen McArthur
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, United States
| | - Noel T Mueller
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, United States
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO 80045, United States
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| |
Collapse
|
4
|
Haj-Husein I, Kubow S, Koski KG. Untargeted Lipidomic Profiling of Amniotic Fluid Reveals Dysregulated Lipid Metabolism in Healthy Normal-Weight Mothers with Fetal Macrosomia. Nutrients 2024; 16:3804. [PMID: 39599591 PMCID: PMC11597394 DOI: 10.3390/nu16223804] [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: 09/23/2024] [Revised: 10/30/2024] [Accepted: 11/02/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Alterations in maternal lipid metabolism have been elucidated by several studies in relation to macrosomia. However, the lipidome of the intrauterine compartment associated with macrosomia, particularly in early pregnancy, remains largely unknown. OBJECTIVES (1) To compare the lipidomic profile of early 2nd trimester amniotic fluid (AF) of healthy mothers with normal body mass index who gave birth to large-for-gestational age (LGA) versus appropriate-for-gestational age (AGA) infants; and (2) to examine if insulin and glucose concentrations in AF were associated with the AF lipidomic profile. METHODS In this nested case-control study, bio-banked AF samples were collected from pregnant women undergoing routine amniocentesis at 12-22 weeks of gestation. A subsample of 15 LGA infants (cases) were contrasted with 15 AGA infants (controls). An untargeted lipidomics analysis using liquid chromatography quadrupole time-of-flight mass spectrometry was conducted. Univariate and multivariate statistical analyses (principal component analysis and partial least-squares discriminant analysis) were used to extract differentially abundant (DA) features with high variable importance in projection (VIP) scores. RESULTS LGA AF was characterized by elevations of 30 phosphatidic acid species. Among other DA features, sphingomyelin (SM 14:0;O2/20:1) had the highest VIP score and was markedly elevated in LGA AF. Neither insulin nor glucose was associated with 2nd trimester AF lipidomic profiles in these healthy, normal-weight mothers. CONCLUSION These findings provide evidence of early dysregulated lipid metabolism in healthy, normal-weight mothers with LGA infants.
Collapse
Affiliation(s)
- Isra’a Haj-Husein
- School of Human Nutrition, McGill University, Ste-Anne de Bellevue, QC H9X 3V9, Canada; (S.K.); (K.G.K.)
| | | | | |
Collapse
|
5
|
Mizuno S, Nagaie S, Sugawara J, Tamiya G, Obara T, Ishikuro M, Kuriyama S, Yaegashi N, Tanaka H, Yamamoto M, Ogishima S. Early prediction of hypertensive disorders of pregnancy toward preventive early intervention. AJOG GLOBAL REPORTS 2024; 4:100383. [PMID: 39524694 PMCID: PMC11550347 DOI: 10.1016/j.xagr.2024.100383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Background Various disease prediction models have been developed, capitalizing on the wide use of electronic health records, but environmental factors that are important in the development of noncommunicable diseases are rarely included in the prediction models. Hypertensive disorders of pregnancy are leading causes of maternal morbidity and mortality and are known to cause several serious complications later in life. Objective This study aims to develop early hypertensive disorders of pregnancy prediction models using comprehensive environmental factors based on self-report questionnaires in early pregnancy. Study Design We developed machine learning and artificial intelligence models for the early prediction of hypertensive disorders of pregnancy using early pregnancy data from approximately 23,000 pregnancies in the Tohoku Medical Megabank Birth and Three Generation Cohort Study. We clarified the important features for prediction based on regression coefficients or Gini coefficients of the interpretable artificial intelligence models (i.e., logistic regression, random forest and XGBoost models) among our developed models. Results The performance of the early hypertensive disorders of pregnancy prediction models reached an area under the receiver operating characteristic curve of 0.93, demonstrating that the early hypertensive disorders of pregnancy prediction models developed in this study retain sufficient performance in hypertensive disorders of pregnancy prediction. Among the early prediction models, the best performing model was based on self-reported questionnaire data in early pregnancy (mean of 20.2 gestational weeks at filling) which consist of comprehensive lifestyles. The interpretation of the models reveals that both eating habits were dominantly important for prediction. Conclusion We have developed high-performance models for early hypertensive disorders of pregnancy prediction using large-scale cohort data from the Tohoku Medical Megabank project. Our study clearly revealed that the use of comprehensive lifestyles from self-report questionnaires led us to predict hypertensive disorders of pregnancy risk at the early stages of pregnancy, which will aid early intervention to reduce the risk of hypertensive disorders of pregnancy.
Collapse
Affiliation(s)
- Satoshi Mizuno
- Department of Informatics for Genomic Medicine, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan (Mizuno, Nagaie, and Ogishima)
| | - Satoshi Nagaie
- Department of Informatics for Genomic Medicine, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan (Mizuno, Nagaie, and Ogishima)
| | - Junichi Sugawara
- Department of Feto-Maternal Medical Science, Tohoku Medical Megabank Organization, Tohoku University. Department of Gynecology and Obstetrics, Tohoku University Graduate School of Medicine, Tohoku University, Miyagi, Japan (Sugawara)
| | - Gen Tamiya
- Department of Statistical Genetics and Genomics, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan (Tamiya)
| | - Taku Obara
- Department of Molecular Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan (Obara, Ishikuro, and Kuriyama)
| | - Mami Ishikuro
- Department of Molecular Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan (Obara, Ishikuro, and Kuriyama)
| | - Shinichi Kuriyama
- Department of Molecular Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan (Obara, Ishikuro, and Kuriyama)
| | - Nobuo Yaegashi
- Department of Gynecology and Obstetrics, Tohoku University Graduate School of Medicine, Tohoku University, Miyagi, Japan (Yaegashi)
| | - Hiroshi Tanaka
- Department of Bioclinical Informatics, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan (Tanaka)
| | - Masayuki Yamamoto
- Department of Integrative Genomics, Tohoku Medical Megabank Organization and Department of Medical Biochemistry, Graduate School of Medicine, Tohoku University, Miyagi, Japan (Yamamoto)
| | - Soichi Ogishima
- Department of Informatics for Genomic Medicine, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan (Mizuno, Nagaie, and Ogishima)
| |
Collapse
|
6
|
Turkoglu O, Citil A, Katar C, Mert I, Quinn RA, Bahado-Singh RO, Graham SF. Untargeted Metabolomic Biomarker Discovery for the Detection of Ectopic Pregnancy. Int J Mol Sci 2024; 25:10333. [PMID: 39408663 PMCID: PMC11476625 DOI: 10.3390/ijms251910333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Ectopic pregnancy (EP) is the leading cause of maternal morbidity and mortality in the first trimester. Using an untargeted metabolomic approach, we sought to identify putative plasma biomarkers using tandem liquid chromatography-mass spectrometry for the detection of tubal EP. This case-control study included the prospective recruitment of 50 tubal EP cases and 50 early intrauterine pregnancy controls. To avoid over-fitting, logistic regression models were developed in a randomly selected discovery group (30 cases vs. 30 controls) and validated in the test group (20 cases vs. 20 controls). In total, 585 mass spectral features were detected, of which 221 molecular features were significantly altered in EP plasma (p < 0.05). Molecular networking and metabolite identification was employed using the Global Natural Products Social Molecular Networking (GNPS) database, which identified 97 metabolites at a high confidence level. Top significant metabolites include subclasses of sphingolipids, carnitines, glycerophosphocholines, and tryptophan metabolism. The top regression model, consisting of D-erythro-sphingosine and oleoyl-carnitine, was validated in a test group and achieved an area under receiving operating curve (AUC) (95% CI) = 0.962 (0.910-1) with a sensitivity of 100% and specificity of 95.9%. Metabolite alterations indicate alterations related to inflammation and abnormal placentation in EP. The validation of these metabolite biomarkers in the future could potentially result in improved early diagnosis.
Collapse
Affiliation(s)
- Onur Turkoglu
- Department of Obstetrics and Gynecology, Maternal Fetal Medicine Division, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ayse Citil
- Department of Obstetrics and Gynecology, Zekai Tahir Burak Women’s Health Education and Research Hospital, Ankara 06230, Turkey
| | - Ceren Katar
- Department of Obstetrics and Gynecology, Zekai Tahir Burak Women’s Health Education and Research Hospital, Ankara 06230, Turkey
| | - Ismail Mert
- Department of Obstetrics and Gynecology, Division of Gynecological Oncology, Advocate Health, Chicago, IL 60642, USA
| | - Robert A. Quinn
- Department of Biochemistry, Michigan State University, Lansing, MI 48824, USA
| | - Ray O. Bahado-Singh
- Department of Obstetrics and Gynecology, Oakland University William Beaumont School of Medicine, Corewell Health, William Beaumont University Hospital, Royal Oak, MI 48073, USA
| | - Stewart F. Graham
- Department of Obstetrics and Gynecology, Oakland University William Beaumont School of Medicine, Corewell Health, William Beaumont University Hospital, Royal Oak, MI 48073, USA
- Metabolomics Department, Corewell Health Research Institute, William Beaumont University Hospital, Royal Oak, MI 48073, USA
| |
Collapse
|
7
|
Youssef L, Testa L, Crovetto F, Crispi F. 10. Role of high dimensional technology in preeclampsia (omics in preeclampsia). Best Pract Res Clin Obstet Gynaecol 2024; 92:102427. [PMID: 37995432 DOI: 10.1016/j.bpobgyn.2023.102427] [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: 06/01/2023] [Revised: 07/05/2023] [Accepted: 08/06/2023] [Indexed: 11/25/2023]
Abstract
Preeclampsia is a pregnancy-specific disease that has no known precise cause. Integrative biology approach based on multi-omics has been applied to identify upstream pathways and better understand the pathophysiology of preeclampsia. At DNA level, genomics and epigenomics studies have revealed numerous genetic variants associated with preeclampsia, including those involved in regulating blood pressure and immune response. Transcriptomics analyses have revealed altered expression of genes in preeclampsia, particularly those related to inflammation and angiogenesis. At protein level, proteomics studies have identified potential biomarkers for preeclampsia diagnosis and prediction in addition to revealing the main pathophysiological pathways involved in this disease. At metabolite level, metabolomics has highlighted altered lipid and amino acid metabolisms in preeclampsia. Finally, microbiomics studies have identified dysbiosis in the gut and vaginal microbiota in pregnant women with preeclampsia. Overall, omics technologies have improved our understanding of the complex molecular mechanisms underlying preeclampsia. However, further research is warranted to fully integrate and translate these omics findings into clinical practice.
Collapse
Affiliation(s)
- Lina Youssef
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, IDIBAPS, University of Barcelona, Barcelona, Spain; Institut de Recerca August Pi Sunyer (IDIBAPS), Barcelona, Spain; Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain.
| | - Lea Testa
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Francesca Crovetto
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, IDIBAPS, University of Barcelona, Barcelona, Spain; Institut de Recerca Sant Joan de Deu (IRSJD), Barcelona, Spain
| | - Fatima Crispi
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, IDIBAPS, University of Barcelona, Barcelona, Spain; Institut de Recerca August Pi Sunyer (IDIBAPS), Barcelona, Spain; Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| |
Collapse
|
8
|
Su AL, Harris SM, Elkin ER, Karnovsky A, Colacino J, Loch-Caruso RK. Trichloroethylene Metabolite S-(1,2-Dichlorovinyl)-l-cysteine Stimulates Changes in Energy Metabolites and Amino Acids in the BeWo Human Placental Trophoblast Model during Syncytialization. Chem Res Toxicol 2023; 36:882-899. [PMID: 37162359 PMCID: PMC10499396 DOI: 10.1021/acs.chemrestox.3c00007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Syncytialization, the fusion of cytotrophoblasts into an epithelial barrier that constitutes the maternal-fetal interface, is a crucial event of placentation. This process is characterized by distinct changes to amino acid and energy metabolism. A metabolite of the industrial solvent trichloroethylene (TCE), S-(1,2-dichlorovinyl)-l-cysteine (DCVC), modifies energy metabolism and amino acid abundance in HTR-8/SVneo extravillous trophoblasts. In the current study, we investigated DCVC-induced changes to energy metabolism and amino acids during forskolin-stimulated syncytialization in BeWo cells, a human villous trophoblastic cell line that models syncytialization in vitro. BeWo cells were exposed to forskolin at 100 μM for 48 h to stimulate syncytialization. During syncytialization, BeWo cells were also treated with DCVC at 0 (control), 10, or 20 μM. Following treatment, the targeted metabolomics platform, "Tricarboxylic Acid Plus", was used to identify changes in energy metabolism and amino acids. DCVC treatment during syncytialization decreased oleic acid, aspartate, proline, uridine diphosphate (UDP), UDP-d-glucose, uridine monophosphate, and cytidine monophosphate relative to forskolin-only treatment controls, but did not increase any measured metabolite. Notable changes stimulated by syncytialization in the absence of DCVC included increased adenosine monophosphate and guanosine monophosphate, as well as decreased aspartate and glutamate. Pathway analysis revealed multiple pathways in amino acid and sugar metabolisms that were altered with forskolin-stimulated syncytialization alone and DCVC treatment during syncytialization. Analysis of ratios of metabolites within the pathways revealed that DCVC exposure during syncytialization changed metabolite ratios in the same or different direction compared to syncytialization alone. Building off our oleic acid findings, we found that extracellular matrix metalloproteinase-2, which is downstream in oleic acid signaling, underwent the same changes as oleic acid. Together, the metabolic changes stimulated by DCVC treatment during syncytialization suggest changes in energy metabolism and amino acid abundance as potential mechanisms by which DCVC could impact syncytialization and pregnancy.
Collapse
Affiliation(s)
- Anthony L. Su
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan 48109
| | - Sean M. Harris
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan 48109
| | - Elana R. Elkin
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan 48109
| | - Alla Karnovsky
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Justin Colacino
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan 48109
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Rita Karen Loch-Caruso
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
9
|
Chen L, Dai J, Fei Z, Liu X, Zhu Y, Rahman ML, Lu R, Mitro SD, Yang J, Hinkle SN, Chen Z, Song Y, Zhang C. Metabolomic biomarkers of the mediterranean diet in pregnant individuals: A prospective study. Clin Nutr 2023; 42:384-393. [PMID: 36753781 PMCID: PMC10029322 DOI: 10.1016/j.clnu.2023.01.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/14/2022] [Accepted: 01/08/2023] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND AIMS Metabolomic profiling is a systematic approach to identifying biomarkers for dietary patterns. Yet, metabolomic markers for dietary patterns in pregnant individuals have not been investigated. The aim of this study was to identify plasma metabolomic markers and metabolite panels that are associated with the Mediterranean diet in pregnant individuals. METHODS This is a prospective study of 186 pregnant individuals who had both dietary intake and metabolomic profiles measured from the Fetal Growth Studies-Singletons cohort. Dietary intakes during the peri-conception/1st trimester and the second trimester were accessed at 8-13 and 16-22 weeks of gestation, respectively. Adherence to the Mediterranean diet was measured by the alternate Mediterranean Diet (aMED) score. Fasting plasma samples were collected at 16-22 weeks and untargeted metabolomics profiling was performed using the mass spectrometry-based platforms. Metabolites individually or jointly associated with aMED scores were identified using linear regression and least absolute shrinkage and selection operator (LASSO) regression models with adjustment for potential confounders, respectively. RESULTS Among 459 annotated metabolites, 64 and 41 were individually associated with the aMED scores of the diet during the peri-conception/1st trimester and during the second trimester, respectively. Fourteen metabolites were associated with the Mediterranean diet in both time windows. Most Mediterranean diet-related metabolites were lipids (e.g., acylcarnitine, cholesteryl esters (CEs), linoleic acid, long-chain triglycerides (TGs), and phosphatidylcholines (PCs), amino acids, and sugar alcohols. LASSO regressions also identified a 10 metabolite-panel that were jointly associated with aMED score of the diet during the peri-conception/1st trimester (AUC: 0.74; 95% CI: 0.57, 0.91) and a 3 metabolites-panel in the 2nd trimester (AUC: 0.68; 95% CI: 0.50, 0.86). CONCLUSION We identified plasma metabolomic markers for the Mediterranean diet among pregnant individuals. Some of them have also been reported in previous studies among non-pregnant populations, whereas others are novel. The results from our study warrant replication in pregnant individuals by future studies. CLINICAL TRIAL REGISTRATION NUMBER This study was registered at ClinicalTrials.gov.
Collapse
Affiliation(s)
- Liwei Chen
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| | - Jin Dai
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| | - Zhe Fei
- Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| | - Xinyue Liu
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| | - Yeyi Zhu
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, 94612, USA.
| | - Mohammad L Rahman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA.
| | - Ruijin Lu
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, USA.
| | - Susanna D Mitro
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, USA.
| | - Jiaxi Yang
- Global Center for Asian Women's Health, and Bia-Echo Asia Centre for Reproductive Longevity & Equality, Yong Loo Lin School of Medicine, National University of Singapore, 117549, Singapore; Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore.
| | - Stefanie N Hinkle
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Zhen Chen
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, USA.
| | - Yiqing Song
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA.
| | - Cuilin Zhang
- Global Center for Asian Women's Health, and Bia-Echo Asia Centre for Reproductive Longevity & Equality, Yong Loo Lin School of Medicine, National University of Singapore, 117549, Singapore; Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore.
| |
Collapse
|
10
|
Enthoven LF, Shi Y, Fay EE, Moreni S, Mao J, Honeyman EM, Smith CK, Whittington D, Brockerhoff SE, Isoherranen N, Totah RA, Hebert MF. The Effects of Pregnancy on Amino Acid Levels and Nitrogen Disposition. Metabolites 2023; 13:242. [PMID: 36837861 PMCID: PMC9961409 DOI: 10.3390/metabo13020242] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/31/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023] Open
Abstract
Limited data are available on the effects of pregnancy on the maternal metabolome. Therefore, the objective of this study was to use metabolomics analysis to determine pathways impacted by pregnancy followed by targeted confirmatory analysis to provide more powerful conclusions about metabolic alterations during pregnancy. Forty-seven pregnant women, 18-50 years of age were included in this study, with each subject serving as their own control. Plasma samples were collected between 25 and 28 weeks gestation and again ≥3 months postpartum for metabolomics analysis utilizing an HILIC/UHPLC/MS/MS assay with confirmatory targeted specific concentration analysis for 10 of the significantly altered amino acids utilizing an LC/MS assay. Principle component analysis (PCA) on metabolomics data clearly separated pregnant and postpartum groups and identified outliers in a preliminary assessment. Of the 980 metabolites recorded, 706 were determined to be significantly different between pregnancy and postpartum. Pathway analysis revealed three significantly impacted pathways, arginine biosynthesis (p = 2 × 10-5 and FDR = 1 × 10-3), valine, leucine, and isoleucine metabolism (p = 2 × 10-5 and FDR = 2 × 10-3), and xanthine metabolism (p = 4 × 10-5 and FDR = 4 × 10-3). Of these we focused analysis on arginine biosynthesis and branched-chain amino acid (BCAA) metabolism due to their clinical importance and interconnected roles in amino acid metabolism. In the confirmational analysis, 7 of 10 metabolites were confirmed as significant and all 10 confirmed the direction of change of concentrations observed in the metabolomics analysis. The data support an alteration in urea nitrogen disposition and amino acid metabolism during pregnancy. These changes could also impact endogenous nitric oxide production and contribute to diseases of pregnancy. This study provides evidence for changes in both the ammonia-urea nitrogen and the BCAA metabolism taking place during pregnancy.
Collapse
Affiliation(s)
- Luke F. Enthoven
- Department of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Yuanyuan Shi
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Emily E. Fay
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Sue Moreni
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Jennie Mao
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Emma M. Honeyman
- Department of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Chase K. Smith
- Department of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Dale Whittington
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA
| | | | - Nina Isoherranen
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA
| | - Rheem A. Totah
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Mary F. Hebert
- Department of Pharmacy, University of Washington, Seattle, WA 98195, USA
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
11
|
Tuytten R, Syngelaki A, Thomas G, Panigassi A, Brown LW, Ortea P, Nicolaides KH. First-trimester preterm preeclampsia prediction with metabolite biomarkers: differential prediction according to maternal body mass index. Am J Obstet Gynecol 2022:S0002-9378(22)02290-6. [PMID: 36539025 DOI: 10.1016/j.ajog.2022.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/08/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Prediction of preeclampsia risk is key to informing effective maternal care. Current screening for preeclampsia at 11 to 13 weeks of gestation using maternal demographic characteristics and medical history with measurements of mean arterial pressure, uterine artery pulsatility index, and serum placental growth factor can identify approximately 75% of women who develop preterm preeclampsia with delivery at <37 weeks of gestation. Further improvements to preeclampsia screening tests will likely require integrating additional biomarkers. Recent research suggests the existence of distinct maternal risk profiles. Therefore, biomarker evaluation should account for the possibility that a biomarker only predicts preeclampsia in a specific maternal phenotype. OBJECTIVE This study aimed to verify metabolite biomarkers as preterm preeclampsia predictors early in pregnancy in all women and across body mass index groups. STUDY DESIGN Observational case-control study drawn from a large prospective study on the early prediction of pregnancy complications in women attending their routine first hospital visit at King's College Hospital, London, United Kingdom, in 2010 to 2015. Pregnant women underwent a complete first-trimester assessment, including the collection of blood samples for biobanking. In 11- to 13-week plasma samples of 2501 singleton pregnancies, the levels of preselected metabolites implicated in the prediction of pregnancy complications were analyzed using a targeted liquid chromatography-mass spectrometry method, yielding high-quality quantification data on 50 metabolites. The ratios of amino acid levels involved in arginine biosynthesis and nitric oxide synthase pathways were added to the list of biomarkers. Placental growth factor and pregnancy-associated plasma protein A were also available for all study subjects, serving as comparator risk predictors. Data on 1635 control and 106 pregnancies complicated by preterm preeclampsia were considered for this analysis, normalized using multiples of medians. Prediction analyses were performed across the following patient strata: all subjects and the body mass index classes of <25, 25 to <30, and ≥30 kg/m2. Adjusted median levels were compared between cases and controls and between each body mass index class group. Odds ratios and 95% confidence intervals were calculated at the mean ±1 standard deviation to gauge clinical prediction merits. RESULTS The levels of 13 metabolites were associated with preterm preeclampsia in the entire study population (P<.05) with particularly significant (P<.01) associations found for 6 of them, namely, 2-hydroxy-(2/3)-methylbutyric acid, 25-hydroxyvitamin D3, 2-hydroxybutyric acid, alanine, dodecanoylcarnitine, and 1-(1Z-octadecenyl)-2-oleoyl-sn-glycero-3-phosphocholine. Fold changes in 7 amino acid ratios, all involving glutamine or ornithine, were also significantly different between cases and controls (P<.01). The predictive performance of some metabolites and ratios differed according to body mass index classification; for example, ornithine (P<.001) and several ornithine-related ratios (P<.0001 to P<.01) were only strongly associated with preterm preeclampsia in the body mass index of <25 kg/m2 group, whereas dodecanoylcarnitine and 3 glutamine ratios were particularly predictive in the body mass index of ≥30 kg/m2 group (P<.01). CONCLUSION Single metabolites and ratios of amino acids related to arginine bioavailability and nitric oxide synthase pathways were associated with preterm preeclampsia risk at 11 to 13 weeks of gestation. Differential prediction was observed according to body mass index classes, supporting the existence of distinct maternal risk profiles. Future studies in preeclampsia prediction should account for the possibility of different maternal risk profiles to improve etiologic and prognostic understanding and, ultimately, clinical utility of screening tests.
Collapse
Affiliation(s)
| | - Argyro Syngelaki
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom
| | | | | | | | | | - Kypros H Nicolaides
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom.
| |
Collapse
|
12
|
Keen B, Cawley A, Reedy B, Fu S. Metabolomics in clinical and forensic toxicology, sports anti-doping and veterinary residues. Drug Test Anal 2022; 14:794-807. [PMID: 35194967 PMCID: PMC9544538 DOI: 10.1002/dta.3245] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 02/13/2022] [Accepted: 02/15/2022] [Indexed: 11/29/2022]
Abstract
Metabolomics is a multidisciplinary field providing workflows for complementary approaches to conventional analytical determinations. It allows for the study of metabolically related groups of compounds or even the study of novel pathways within the biological system. The procedural stages of metabolomics; experimental design, sample preparation, analytical determinations, data processing and statistical analysis, compound identification and validation strategies are explored in this review. The selected approach will depend on the type of study being conducted. Experimental design influences the whole metabolomics workflow and thus needs to be properly assessed to ensure sufficient sample size, minimal introduced and biological variation and appropriate statistical power. Sample preparation needs to be simple, yet potentially global in order to detect as many compounds as possible. Analytical determinations need to be optimised either for the list of targeted compounds or a universal approach. Data processing and statistical analysis approaches vary widely and need to be better harmonised for review and interpretation. This includes validation strategies that are currently deficient in many presented workflows. Common compound identification approaches have been explored in this review. Metabolomics applications are discussed for clinical and forensic toxicology, human and equine sports anti-doping and veterinary residues.
Collapse
Affiliation(s)
- Bethany Keen
- Centre for Forensic ScienceUniversity of Technology SydneyBroadwayNew South WalesAustralia
| | - Adam Cawley
- Australian Racing Forensic LaboratoryRacing NSWSydneyNew South WalesAustralia
| | - Brian Reedy
- School of Mathematical and Physical SciencesUniversity of Technology SydneyBroadwayNew South WalesAustralia
| | - Shanlin Fu
- Centre for Forensic ScienceUniversity of Technology SydneyBroadwayNew South WalesAustralia
| |
Collapse
|
13
|
Mayrink J, Leite DF, Nobrega GM, Costa ML, Cecatti JG. Prediction of pregnancy-related hypertensive disorders using metabolomics: a systematic review. BMJ Open 2022; 12:e054697. [PMID: 35470187 PMCID: PMC9039389 DOI: 10.1136/bmjopen-2021-054697] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 04/05/2022] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE To determine the accuracy of metabolomics in predicting hypertensive disorders in pregnancy. DESIGN Systematic review of observational studies. DATA SOURCES AND STUDY ELIGIBILITY CRITERIA An electronic literature search was performed in June 2019 and February 2022. Two researchers independently selected studies published between 1998 and 2022 on metabolomic techniques applied to predict the condition; subsequently, they extracted data and performed quality assessment. Discrepancies were dealt with a third reviewer. The primary outcome was pre-eclampsia. Cohort or case-control studies were eligible when maternal samples were taken before diagnosis of the hypertensive disorder. STUDY APPRAISAL AND SYNTHESIS METHODS Data on study design, maternal characteristics, how hypertension was diagnosed, metabolomics details and metabolites, and accuracy were independently extracted by two authors. RESULTS Among 4613 initially identified studies on metabolomics, 68 were read in full text and 32 articles were included. Studies were excluded due to duplicated data, study design or lack of identification of metabolites. Metabolomics was applied mainly in the second trimester; the most common technique was liquid-chromatography coupled to mass spectrometry. Among the 122 different metabolites found, there were 23 amino acids and 21 fatty acids. Most of the metabolites were involved with ammonia recycling; amino acid metabolism; arachidonic acid metabolism; lipid transport, metabolism and peroxidation; fatty acid metabolism; cell signalling; galactose metabolism; nucleotide sugars metabolism; lactose degradation; and glycerolipid metabolism. Only citrate was a common metabolite for prediction of early-onset and late-onset pre-eclampsia. Vitamin D was the only metabolite in common for pre-eclampsia and gestational hypertension prediction. Meta-analysis was not performed due to lack of appropriate standardised data. CONCLUSIONS AND IMPLICATIONS Metabolite signatures may contribute to further insights into the pathogenesis of pre-eclampsia and support screening tests. Nevertheless, it is mandatory to validate such methods in larger studies with a heterogeneous population to ascertain the potential for their use in clinical practice. PROSPERO REGISTRATION NUMBER CRD42018097409.
Collapse
Affiliation(s)
- Jussara Mayrink
- Department of Obstetrics and Gynecology, State University of Campinas Faculty of Medical Sciences, Campinas, Brazil
| | - Debora F Leite
- Department of Obstetrics and Gynecology, State University of Campinas Faculty of Medical Sciences, Campinas, Brazil
| | - Guilherme M Nobrega
- Department of Obstetrics and Gynecology, State University of Campinas Faculty of Medical Sciences, Campinas, Brazil
| | - Maria Laura Costa
- Department of Obstetrics and Gynecology, State University of Campinas Faculty of Medical Sciences, Campinas, Brazil
| | - Jose Guilherme Cecatti
- Department of Obstetrics and Gynecology, State University of Campinas Faculty of Medical Sciences, Campinas, Brazil
| |
Collapse
|
14
|
Su AL, Harris SM, Elkin ER, Karnovsky A, Colacino JA, Loch-Caruso R. Trichloroethylene modifies energy metabolites in the amniotic fluid of Wistar rats. Reprod Toxicol 2022; 109:80-92. [PMID: 35301063 PMCID: PMC9000924 DOI: 10.1016/j.reprotox.2022.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 03/05/2022] [Accepted: 03/11/2022] [Indexed: 10/18/2022]
Abstract
Exposure to trichloroethylene (TCE), an industrial solvent, is associated with several adverse pregnancy outcomes in humans and decreased fetal weight in rats. However, effects of TCE on energy metabolites in amniotic fluid, which have associations with pregnancy outcomes, has not been published previously. In the current exploratory study, timed-pregnant Wistar rats were exposed to 480 mg TCE/kg/day via vanilla wafer or to vehicle (wafer) alone from gestational day (GD) 6-16. Amniotic fluid collected on GD 16 was analyzed for metabolites important in energy metabolism using short chain fatty acid and tricarboxylic acid plus platforms (N = 4 samples/sex/treatment). TCE decreased concentrations of the following metabolites in amniotic fluid for both fetal sexes: 6-phosphogluconate, guanosine diphosphate, adenosine diphosphate, adenosine triphosphate, and flavin adenine dinucleotide. TCE decreased fructose 1,6-bisphosphate and guanosine triphosphate concentrations in amniotic fluid of male but not female fetuses. Moreover, TCE decreased uridine diphosphate-D-glucuronate concentrations, and increased arginine and phosphocreatine concentrations, in amniotic fluid of female fetuses only. No metabolites were increased in amniotic fluid of male fetuses. Pathway analysis suggested that TCE altered folate biosynthesis and pentose phosphate pathway in both sexes. Using metabolite ratios to investigate changes within specific pathways, some ratio alterations, including those in arginine metabolism and phenylalanine metabolism, were detected in females only. Ratio analysis also suggested enzymes, including gluconokinase, as potential TCE targets. Together, results from this exploratory study suggest that TCE differentially modified energy metabolites in amniotic fluid based on sex. These findings may inform future studies of TCE reproductive toxicity.
Collapse
Affiliation(s)
- Anthony L Su
- Department of Environmental Health Sciences, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109, USA.
| | - Sean M Harris
- Department of Environmental Health Sciences, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109, USA.
| | - Elana R Elkin
- Department of Environmental Health Sciences, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109, USA.
| | - Alla Karnovsky
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Palmer Commons, 100 Washtenaw Ave #2017, Ann Arbor, MI 48109, USA.
| | - Justin A Colacino
- Department of Environmental Health Sciences, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109, USA; Department of Nutritional Sciences, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109, USA.
| | - Rita Loch-Caruso
- Department of Environmental Health Sciences, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109, USA.
| |
Collapse
|
15
|
Yao M, Xiao Y, Yang Z, Ge W, Liang F, Teng H, Gu Y, Yin J. Identification of Biomarkers for Preeclampsia Based on Metabolomics. Clin Epidemiol 2022; 14:337-360. [PMID: 35342309 PMCID: PMC8943653 DOI: 10.2147/clep.s353019] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/25/2022] [Indexed: 01/15/2023] Open
Abstract
Background Preeclampsia (PE) is a significant cause of maternal and neonatal morbidity and mortality worldwide. However, the pathogenesis of PE is unclear and reliable early diagnostic methods are still lacking. The purpose of this review is to summarize potential metabolic biomarkers and pathways of PE, which might facilitate risk prediction and clinical diagnosis, and obtain a better understanding of specific metabolic mechanisms of PE. Methods This review included human metabolomics studies related to PE in the PubMed, Google Scholar, and Web of Science databases from January 2000 to November 2021. The reported metabolic biomarkers were systematically examined and compared. Pathway analysis was conducted through the online software MetaboAnalyst 5.0. Results Forty-one human studies were included in this systematic review. Several metabolites, such as creatinine, glycine, L-isoleucine, and glucose and biomarkers with consistent trends (decanoylcarnitine, 3-hydroxyisovaleric acid, and octenoylcarnitine), were frequently reported. In addition, eight amino acid metabolism-related, three carbohydrate metabolism-related, one translation-related and one lipid metabolism-related pathways were identified. These biomarkers and pathways, closely related to renal dysfunction, insulin resistance, lipid metabolism disorder, activated inflammation, and impaired nitric oxide production, were very likely to contribute to the progression of PE. Conclusion This study summarized several metabolites and metabolic pathways, which may be associated with PE. These high-frequency differential metabolites are promising to be biomarkers of PE for early diagnosis, and the prominent metabolic pathway may provide new insights for the understanding of the pathogenesis of PE.
Collapse
Affiliation(s)
- Mengxin Yao
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, People’s Republic of China
| | - Yue Xiao
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, People’s Republic of China
| | - Zhuoqiao Yang
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, People’s Republic of China
| | - Wenxin Ge
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, People’s Republic of China
| | - Fei Liang
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, People’s Republic of China
| | - Haoyue Teng
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, People’s Republic of China
| | - Yingjie Gu
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Jieyun Yin
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, People’s Republic of China
- Correspondence: Jieyun Yin, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, People’s Republic of China, Tel/Fax +86 0512 6588036, Email
| |
Collapse
|
16
|
Chaemsaithong P, Sahota DS, Poon LC. First trimester preeclampsia screening and prediction. Am J Obstet Gynecol 2022; 226:S1071-S1097.e2. [PMID: 32682859 DOI: 10.1016/j.ajog.2020.07.020] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/30/2020] [Accepted: 07/14/2020] [Indexed: 12/16/2022]
Abstract
Preeclampsia is a major cause of maternal and perinatal morbidity and mortality. Early-onset disease requiring preterm delivery is associated with a higher risk of complications in both mothers and babies. Evidence suggests that the administration of low-dose aspirin initiated before 16 weeks' gestation significantly reduces the rate of preterm preeclampsia. Therefore, it is important to identify pregnant women at risk of developing preeclampsia during the first trimester of pregnancy, thus allowing timely therapeutic intervention. Several professional organizations such as the American College of Obstetricians and Gynecologists (ACOG) and National Institute for Health and Care Excellence (NICE) have proposed screening for preeclampsia based on maternal risk factors. The approach recommended by ACOG and NICE essentially treats each risk factor as a separate screening test with additive detection rate and screen-positive rate. Evidence has shown that preeclampsia screening based on the NICE and ACOG approach has suboptimal performance, as the NICE recommendation only achieves detection rates of 41% and 34%, with a 10% false-positive rate, for preterm and term preeclampsia, respectively. Screening based on the 2013 ACOG recommendation can only achieve detection rates of 5% and 2% for preterm and term preeclampsia, respectively, with a 0.2% false-positive rate. Various first trimester prediction models have been developed. Most of them have not undergone or failed external validation. However, it is worthy of note that the Fetal Medicine Foundation (FMF) first trimester prediction model (namely the triple test), which consists of a combination of maternal factors and measurements of mean arterial pressure, uterine artery pulsatility index, and serum placental growth factor, has undergone successful internal and external validation. The FMF triple test has detection rates of 90% and 75% for the prediction of early and preterm preeclampsia, respectively, with a 10% false-positive rate. Such performance of screening is superior to that of the traditional method by maternal risk factors alone. The use of the FMF prediction model, followed by the administration of low-dose aspirin, has been shown to reduce the rate of preterm preeclampsia by 62%. The number needed to screen to prevent 1 case of preterm preeclampsia by the FMF triple test is 250. The key to maintaining optimal screening performance is to establish standardized protocols for biomarker measurements and regular biomarker quality assessment, as inaccurate measurement can affect screening performance. Tools frequently used to assess quality control include the cumulative sum and target plot. Cumulative sum is a sensitive method to detect small shifts over time, and point of shift can be easily identified. Target plot is a tool to evaluate deviation from the expected multiple of median and the expected median of standard deviation. Target plot is easy to interpret and visualize. However, it is insensitive to detecting small deviations. Adherence to well-defined protocols for the measurements of mean arterial pressure, uterine artery pulsatility index, and placental growth factor is required. This article summarizes the existing literature on the different methods, recommendations by professional organizations, quality assessment of different components of risk assessment, and clinical implementation of the first trimester screening for preeclampsia.
Collapse
|
17
|
Para R, Romero R, Gomez-Lopez N, Tarca AL, Panaitescu B, Done B, Hsu R, Pacora P, Hsu CD. Maternal circulating concentrations of soluble Fas and Elabela in early- and late-onset preeclampsia. J Matern Fetal Neonatal Med 2022; 35:316-329. [PMID: 32008387 PMCID: PMC10544759 DOI: 10.1080/14767058.2020.1716720] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/17/2019] [Accepted: 01/13/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The Fas/Fas ligand (FASL) system and Elabela-apelin receptor signaling pathways are implicated in the pathophysiology of preeclampsia. The aim of the current study was to investigate whether a model combining the measurement of sFas and Elabela in the maternal circulation may serve as a clinical biomarker for early- and/or late-onset preeclampsia more effectively than measures of each biomarker individually. METHODS Blood samples were collected from 214 women in the following groups: (1) normal pregnancy sampled <34 weeks of gestation (n = 56); (2) patients who developed early-onset preeclampsia (n = 54); (3) normal pregnancy sampled ≥34 weeks of gestation (n = 52); (4) patients who developed late-onset preeclampsia (n = 52). Maternal circulating soluble Fas and Elabela concentrations were determined using sensitive and validated immunoassays. Two sample t-tests, multivariate logistic regression, and receiver operating characteristic curves were used for analyses. RESULTS (1) Women with early-onset preeclampsia, and those with late-onset preeclampsia with placental lesions of maternal vascular malperfusion, had increased concentrations of sFas compared to their gestational age-matched normal controls; (2) women with late-onset preeclampsia, but not those with early-onset preeclampsia, had increased concentrations of Elabela compared to their gestational age-matched counterparts; and (3) an increase in both Elabela and sFas concentrations was more strongly associated with late-onset preeclampsia than early-onset preeclampsia relative to models including either of the markers alone. CONCLUSIONS A combined model of maternal sFas and Elabela concentrations provides a stronger association with late-onset preeclampsia than either protein alone. This finding demonstrates the possibility to improve the classification of late-onset preeclampsia by combining the results of both molecular biomarkers.
Collapse
Affiliation(s)
- Robert Para
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Florida International University, Miami, Florida, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Adi L. Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, USA
| | - Bogdan Panaitescu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Bogdan Done
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Richard Hsu
- Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Percy Pacora
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Chaur-Dong Hsu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
18
|
C1431T Variant of PPARγ Is Associated with Preeclampsia in Pregnant Women. Life (Basel) 2021; 11:life11101052. [PMID: 34685423 PMCID: PMC8540421 DOI: 10.3390/life11101052] [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: 08/13/2021] [Revised: 10/01/2021] [Accepted: 10/02/2021] [Indexed: 12/16/2022] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is essential for placental development, whose SNPs have shown increased susceptibility to pregnancy-related diseases, such as preeclampsia. Our aim was to investigate the association between preeclampsia and three PPARγ SNPs (Pro12Ala, C1431T, and C681G), which together with nine clinical factors were used to build a pragmatic model for preeclampsia prediction. Data were collected from 1648 women from the EDEN cohort, of which 35 women had preeclamptic pregnancies, and the remaining 1613 women had normal pregnancies. Univariate analysis comparing preeclamptic patients to the control resulted in the SNP C1431T being the only factor significantly associated with preeclampsia (p < 0.05), with a confidence interval of 95% and odds ratio ranging from 4.90 to 8.75. On the other hand, three methods of multivariate feature selection highlighted seven features that could be potential predictors of preeclampsia: maternal C1431T and C681G variants, obesity, body mass index, number of pregnancies, primiparity, cigarette use, and education. These seven features were further used as input into eight different machine-learning algorithms to create predictive models, whose performances were evaluated based on metrics of accuracy and the area under the receiver operating characteristic curve (AUC). The boost tree-based model performed the best, with respective accuracy and AUC values of 0.971 ± 0.002 and 0.991 ± 0.001 in the training set and 0.951 and 0.701 in the testing set. A flowchart based on the boost tree model was constructed to depict the procedure for preeclampsia prediction. This final decision tree showed that the C1431T variant of PPARγ is significantly associated with susceptibility to preeclampsia. We believe that this final decision tree could be applied in the clinical prediction of preeclampsia in the very early stages of pregnancy.
Collapse
|
19
|
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
|
20
|
Rodríguez KBM, Gómez LMR, Yáñez LC, Ramírez RF, Ornelas-Rebolledo O, Borjas-García JA, Pérez-Vázquez F, Aguilar MR. Application of the Electronic Nose in Predicting Preeclampsia in High-risk Pregnancies. Pilot Study. Arch Med Res 2021; 52:561-568. [PMID: 33597111 DOI: 10.1016/j.arcmed.2021.01.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/16/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Preeclampsia is a syndrome that affects 2-8 % of pregnancies worldwide and is the leading cause of maternal death. Therefore, early detection is crucial to identify women who require clinical monitoring during pregnancy and to evaluate new preventive therapies before clinical symptoms occur. METHODS The chemical fingerprints of the urine from three study groups pregnant with Preeclampsia, Healthy Pregnant (HP) and pregnant at High Risk of Preeclampsia (HRP) were evaluated using an electronic nose and the data obtained were subjected to principal component analysis (PCA), Canonical Analysis of Principal Coordinates (CAP), Partial Least Squares - Discriminant Analysis (PLS-DA) and ROC curves to determine the diagnostic power of the test. RESULTS A separation was found between the patients with preeclampsia and HP explaining 99% of the variability of the data. Subsequently, a CAP was obtained with a correct classification of 100%, and the PLS-DA was obtained an accuracy of 88%. With the results of axis CAP1, a ROC curve was performed resulting in a sensitivity of 100% and a specificity of 95.5%. Based on the CAP model it was found that 36% (n=9) of the HRP patients would develop preeclampsia based on the metabolites found in urine. CONCLUSION metabolomics can be used as a tool for early detection of preeclampsia in high-risk pregnant women, using portable olfactory technology.
Collapse
Affiliation(s)
- Karen Beatriz Méndez Rodríguez
- Centro de Investigación Aplicada en Ambiente y Salud, CIACYT, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, México
| | - Luis Manuel Ramírez Gómez
- Departamento de Nefrología, Hospital Central, Dr. Ignacio Morones Prieto, San Luis Potosí, San Luis Potosí, México
| | - Leticia Carrizales Yáñez
- Centro de Investigación Aplicada en Ambiente y Salud, CIACYT, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, México
| | - Rogelio Flores Ramírez
- CONACYT, Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, México
| | - Omar Ornelas-Rebolledo
- Centro Labinnova de Investigación en Respiración para la detección temprana de enfermedades, Guadalajara, México
| | - Jaime Antonio Borjas-García
- Departamento de Nefrología, Hospital Central, Dr. Ignacio Morones Prieto, San Luis Potosí, San Luis Potosí, México
| | - Francisco Pérez-Vázquez
- CONACYT, Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, México
| | - Maribel Rodríguez Aguilar
- Centro de Investigación Aplicada en Ambiente y Salud, CIACYT, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, México; Centro Labinnova de Investigación en Respiración para la detección temprana de enfermedades, Guadalajara, México.
| |
Collapse
|
21
|
Harville EW, Li YY, Pan K, McRitchie S, Pathmasiri W, Sumner S. Untargeted analysis of first trimester serum to reveal biomarkers of pregnancy complications: a case-control discovery phase study. Sci Rep 2021; 11:3468. [PMID: 33568690 PMCID: PMC7876105 DOI: 10.1038/s41598-021-82804-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 01/15/2021] [Indexed: 11/23/2022] Open
Abstract
Understanding of causal biology and predictive biomarkers are lacking for hypertensive disorders of pregnancy (HDP) and preterm birth (PTB). First-trimester serum specimens from 51 cases of HDP, including 18 cases of pre-eclampsia (PE) and 33 cases of gestational hypertension (GH); 53 cases of PTB; and 109 controls were obtained from the Global Alliance to Prevent Prematurity and Stillbirth repository. Metabotyping was conducted using liquid chromatography high resolution mass spectroscopy and nuclear magnetic resonance spectroscopy. Multivariable logistic regression was used to identify signals that differed between groups after controlling for confounders. Signals important to predicting HDP and PTB were matched to an in-house physical standards library and public databases. Pathway analysis was conducted using GeneGo MetaCore. Over 400 signals for endogenous and exogenous metabolites that differentiated cases and controls were identified or annotated, and models that included these signals produced substantial improvements in predictive power beyond models that only included known risk factors. Perturbations of the aminoacyl-tRNA biosynthesis, L-threonine, and renal secretion of organic electrolytes pathways were associated with both HDP and PTB, while pathways related to cholesterol transport and metabolism were associated with HDP. This untargeted metabolomics analysis identified signals and common pathways associated with pregnancy complications.
Collapse
Affiliation(s)
- E W Harville
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, Epidemiology #8318, 1440 Canal St. Ste. 2001, New Orleans, LA, 70112, USA.
| | - Y-Y Li
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill School of Public Health, CB#74612, Chapel Hill, NC, 27599-7461, USA
| | - K Pan
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, Epidemiology #8318, 1440 Canal St. Ste. 2001, New Orleans, LA, 70112, USA
| | - S McRitchie
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill School of Public Health, CB#74612, Chapel Hill, NC, 27599-7461, USA
| | - W Pathmasiri
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill School of Public Health, CB#74612, Chapel Hill, NC, 27599-7461, USA
| | - S Sumner
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill School of Public Health, CB#74612, Chapel Hill, NC, 27599-7461, USA.
| |
Collapse
|
22
|
Liu L, Liu L, Wang J, Zheng Q, Jin B, Sun L. Differentiation of gestational diabetes mellitus by nuclear magnetic resonance-based metabolic plasma analysis. J Biomed Res 2021; 35:351-360. [PMID: 34511531 PMCID: PMC8502693 DOI: 10.7555/jbr.35.20200191] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
This study aimed to investigate the metabolic profile of gestational diabetes mellitus (GDM) at both antepartum and postpartum periods. Seventy pregnant women were divided into three groups: the normal glucose-tolerant group (NGT, n=35), the abnormal glucose-tolerant groups without insulin therapy (A1GDM, n=24) or with insulin therapy (A2GDM, n=11). Metabolic profiles of the plasma were acquired by proton nuclear magnetic resonance (1H-NMR) spectroscopy and analyzed by multivariate statistical data analysis. The relationship between demographic parameters and the potential metabolite biomarkers was further explored. Group antepartum or postpartum showed similar metabolic trends. Compare with those of the NGT group, the levels of 2-hydroxybutyrate, lysine, acetate, glutamine, succinate, tyrosine, formate, and all three BCAAs (leucine, valine, isoleucine) in the A2GDM group were increased dramatically, and the levels of lysine, acetate, and formate in the A1GDM group were elevated significantly. The dramatically decreased levels of 3-methyl-2-oxovalerate and methanol were observed both in the A1GDM group and A2GDM group. Compare to the A1GDM group, the branched-chain amino acids (BCAAs) of leucine, valine, and isoleucine were increased dramatically in the A2GDM group. The levels of aromatic amino acids (AAAs), tyrosine and phenylalanine, were significantly increased in GDM women, consistent with the severity of GDM. Interference of amino acid metabolism and disturbance in energy metabolism occurred in women with different grades of GDM. Metabolic profiles could reflect the severity of GDM. Plasma BCAA concentrations showing strong positive correlations with weight and pre-delivery BMI. This study provides a new perspective to understand the pathogenesis and etiology of GDM, which may help the clinical management and treatment of GDM.
Collapse
Affiliation(s)
- Liping Liu
- Department of Obstetrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Lenan Liu
- Department of Obstetrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Junsong Wang
- Center for Molecular Metabolism, Nanjing University of Science and Technology, Nanjing, Jiangsu 210094, China
| | - Qi Zheng
- Center for Molecular Metabolism, Nanjing University of Science and Technology, Nanjing, Jiangsu 210094, China
| | - Bai Jin
- Department of Obstetrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Lizhou Sun
- Department of Obstetrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
23
|
Wang H, Ma L, Pan X, Du Z, Chen Y. Novel associations of SNPs MYLIP rs3757354 and ABCA1 2230806 gene with early-onset-preeclampsia: A case-control candidate genetic study. Pregnancy Hypertens 2021; 23:185-190. [PMID: 33450693 DOI: 10.1016/j.preghy.2020.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To investigate the association between MYLIP rs3757354 and ABCA1 2230806 single nucleotide polymorphisms in women with preeclampsia in China. STUDY DESIGN The case-control study involved 205 patients with preeclampsia and 145 controls. All women with preeclampsia were divided into two groups: 78 patients with early-onset preeclampsia and 127 with late-onset preeclampsia. MAIN OUTCOME MEASURE MYLIP rs3757354 and ABCA1 rs2230806 SNPs were analyzed through multiplex PCR for targeted next-generation sequencing technology. A secondary outcome was lipid profile changes and liver function in women with PE. RESULTS Maternal age (OR: 1.073, 95% CI = 1.006-1.145), BMI (OR: 1.118, 95% CI = 1.040-1.201), TG/HDL-C (OR: 1.536, 95% CI = 1.080-2.183), and TT genotype of SNP rs3757354 (OR: 3.238, 95% CI = 1.313-7.990) were associated with EOPE risk. Our study found that patients with TT genotype of ABCA1 rs2230806 had more severe hepatic dysfunction and higher HDL levels in the EOPE group compared with CC/CT genotype. There was no association between rs2230806 and the risk of PE. CONCLUSION The polymorphisms of rs3757354 are associated with the risk of EOPE in Chinese pregnant women. The TT genotype in ABCA1 rs2230806 is a strong predictive risk for elevated aminotransferase levels in pregnant women with EOPE.
Collapse
Affiliation(s)
- He Wang
- The First Hospital of Jilin University, Department of Obstetrics, Changchun, Jilin Province 130021, China
| | - Lingyu Ma
- The First Hospital of Jilin University, Department of Obstetrics, Changchun, Jilin Province 130021, China
| | - Xuefeng Pan
- The First Hospital of Jilin University, Department of Obstetrics, Changchun, Jilin Province 130021, China
| | - Zhaoli Du
- Institute of Genetic Technology, Yinfeng Bilogical Group, Yinfeng Gene Technology Company Limited, Jinan, Shandong Province 250014, China
| | - Ying Chen
- The First Hospital of Jilin University, Department of Obstetrics, Changchun, Jilin Province 130021, China.
| |
Collapse
|
24
|
Lee SM, Kang Y, Lee EM, Jung YM, Hong S, Park SJ, Park CW, Norwitz ER, Lee DY, Park JS. Metabolomic biomarkers in midtrimester maternal plasma can accurately predict the development of preeclampsia. Sci Rep 2020; 10:16142. [PMID: 32999354 PMCID: PMC7527521 DOI: 10.1038/s41598-020-72852-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 09/01/2020] [Indexed: 01/08/2023] Open
Abstract
Early identification of patients at risk of developing preeclampsia (PE) would allow providers to tailor their prenatal management and adopt preventive strategies, such as low-dose aspirin. Nevertheless, no mid-trimester biomarkers have as yet been proven useful for prediction of PE. This study investigates the ability of metabolomic biomarkers in mid-trimester maternal plasma to predict PE. A case–control study was conducted including 33 pregnant women with mid-trimester maternal plasma (gestational age [GA], 16–24 weeks) who subsequently developed PE and 66 GA-matched controls with normal outcomes (mid-trimester cohort). Plasma samples were comprehensively profiled for primary metabolic and lipidomic signatures based on gas chromatography time-of-flight mass spectrometry (GC-TOF MS) and liquid chromatography Orbitrap mass spectrometry (LC-Orbitrap MS). A potential biomarker panel was computed based on binary logistic regression and evaluated using receiver operating characteristic (ROC) analysis. To evaluate whether this panel can be also used in late pregnancy, a retrospective cohort study was conducted using plasma collected from women who delivered in the late preterm period because of PE (n = 13) or other causes (n = 21) (at-delivery cohort). Metabolomic biomarkers were compared according to the indication for delivery. Performance of the metabolomic panel to identify patients with PE was compared also to a commonly used standard, the plasma soluble fms-like tyrosine kinase-1/placental growth factor (sFlt-1/PlGF) ratio. In the mid-trimester cohort, a total of 329 metabolites were identified and semi-quantified in maternal plasma using GC-TOF MS and LC-Orbitrap-MS. Binary logistic regression analysis proposed a mid-trimester biomarker panel for the prediction of PE with five metabolites (SM C28:1, SM C30:1, LysoPC C19:0, LysoPE C20:0, propane-1,3-diol). This metabolomic model predicted PE better than PlGF (AUC [95% CI]: 0.868 [0.844–0.891] vs 0.604 [0.485–0.723]) and sFlt-1/PlGF ratio. Analysis of plasma from the at-delivery cohort confirmed the ability of this biomarker panel to distinguish PE from non-PE, with comparable discrimination power to that of the sFlt-1/PlGF ratio. In conclusion, an integrative metabolomic biomarker panel in mid-trimester maternal plasma can accurately predict the development of PE and showed good discriminatory power in patients with PE at delivery.
Collapse
Affiliation(s)
- Seung Mi Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Yujin Kang
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute for Agricultural and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Eun Mi Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute for Agricultural and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Young Mi Jung
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Subeen Hong
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Soo Jin Park
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute for Agricultural and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Chan-Wook Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Errol R Norwitz
- Department of Obstetrics and Gynecology, Tufts University School of Medicine, Boston, MA, USA
| | - Do Yup Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute for Agricultural and Life Sciences, Seoul National University, Seoul, 08826, Korea.
| | - Joong Shin Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, 03080, Korea.
| |
Collapse
|
25
|
Sovio U, McBride N, Wood AM, Masconi KL, Cook E, Gaccioli F, Charnock-Jones DS, Lawlor DA, Smith GCS. 4-Hydroxyglutamate is a novel predictor of pre-eclampsia. Int J Epidemiol 2020; 49:301-311. [PMID: 31098639 PMCID: PMC7124498 DOI: 10.1093/ije/dyz098] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2019] [Indexed: 11/28/2022] Open
Abstract
Background Pre-term pre-eclampsia is a major cause of maternal and perinatal morbidity and mortality worldwide. A multi-centre randomized–controlled trial has shown that first-trimester screening followed by treatment of high-risk women with aspirin reduces the risk of pre-term pre-eclampsia. However, the biomarkers currently employed in risk prediction are only weakly associated with the outcome. Methods We conducted a case–cohort study within the Pregnancy Outcome Prediction study to analyse untargeted maternal serum metabolomics in samples from 12, 20, 28 and 36 weeks of gestational age (wkGA) in women with pre-eclampsia delivering at term (n = 165) and pre-term (n = 29), plus a random sample of the cohort (n = 325). We used longitudinal linear mixed models to identify candidate metabolites at 20/28 wkGA that differed by term pre-eclampsia status. Candidates were validated using measurements at 36 wkGA in the same women. We then tested the association between the 12-, 20- and 28-wkGA measurements and pre-term pre-eclampsia. We externally validated the association using 24- to 28-wkGA samples from the Born in Bradford study (25 cases and 953 controls). Results We identified 100 metabolites that differed most at 20/28 wkGA in term pre-eclampsia. Thirty-three of these were validated (P < 0.0005) at 36 wkGA. 4-Hydroxyglutamate and C-glycosyltryptophan were independently predictive at 36 wkGA of term pre-eclampsia. 4-Hydroxyglutamate was also predictive (area under the receiver operating characteristic curve, 95% confidence interval) of pre-term pre-eclampsia at 12 (0.673, 0.558–0.787), 20 (0.731, 0.657–0.806) and 28 wkGA (0.733, 0.627–0.839). The predictive ability of 4-hydroxyglutamate at 12 wkGA was stronger than two existing protein biomarkers, namely PAPP-A (0.567, 0.439–0.695) and placenta growth factor (0.589, 0.463–0.714). Finally, 4-hydroxyglutamate at 24–28 wkGA was positively associated with pre-eclampsia (term or pre-term) among women from the Born in Bradford study. Conclusions 4-hydroxyglutamate is a novel biochemical predictor of pre-eclampsia that provides better first-trimester prediction of pre-term disease than currently employed protein biomarkers.
Collapse
Affiliation(s)
- Ulla Sovio
- Department of Obstetrics and Gynaecology, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge, UK.,Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Nancy McBride
- NIHR Bristol Biomedical Research Centre, Bristol, UK.,MRC Integrative Epidemiology Unit, at the University of Bristol, Bristol, UK.,Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Angela M Wood
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Katya L Masconi
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Emma Cook
- Department of Obstetrics and Gynaecology, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Francesca Gaccioli
- Department of Obstetrics and Gynaecology, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge, UK.,Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - D Stephen Charnock-Jones
- Department of Obstetrics and Gynaecology, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge, UK.,Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Debbie A Lawlor
- NIHR Bristol Biomedical Research Centre, Bristol, UK.,MRC Integrative Epidemiology Unit, at the University of Bristol, Bristol, UK.,Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Gordon C S Smith
- Department of Obstetrics and Gynaecology, University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge, UK.,Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
26
|
Tan B, Ma Y, Zhang L, Li N, Zhang J. The application of metabolomics analysis in the research of gestational diabetes mellitus and preeclampsia. J Obstet Gynaecol Res 2020; 46:1310-1318. [PMID: 32500661 DOI: 10.1111/jog.14304] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/26/2020] [Accepted: 04/30/2020] [Indexed: 12/14/2022]
Abstract
AIM The aim of the study was to investigate the difference of the serum metabolic profile between gestational diabetes mellitus (GDM) patients and preeclampsia (PE) patients, to establish the disease differentiation model and to find characteristic metabolites, in order to provide a new idea for the occurrence, development and treatment of the disease. METHODS Twenty-nine patients with GDM group and 29 PE group who were examined in Tianjin No. 3 Central Hospital from March 2018 to August 2018 were enrolled as case group, and 29 normal pregnant women were selected as control group. All the serum samples were analyzed by using the ultra-performance liquid chromatography and mass spectrometry. Based on the multivariate statistical analysis method of pattern recognition, we screened out and identified the differential characteristic metabolites. RESULTS The serum metabolic profile model of GDM group and PE group was successfully constructed. A total of nine characteristic metabolites were screened and identified in this study, including LPC 18:0, LPC 22:6, LPC 16:0, (S)-14-methylhexadecanoic acid, behenic acid, palmitic acid, sphingosine, phytosphingosine and 1,25-dihydroxyvitamin D3-26,23-lactone. Among them, six characteristic metabolites which were LPC 18:0, LPC 22:6, behenic acid, palmitic acid, sphingosine and 1,25-dihydroxyvitamin D3-26,23-lactone all had a significant statistical difference among GDM, PE and normal pregnancy groups (P < 0.05). CONCLUSION The construction of metabolic profile discriminant model has a strong ability to differentiate GDM patients from PE pregnant women. The screened characteristic metabolites can early reflect the disorder of lipid, calcium and phosphorus metabolism of patients, and provide reference and help for the discussion of the occurrence, development and treatment of diseases.
Collapse
Affiliation(s)
- Bing Tan
- Medical Laboratory Department, Tianjin Third Central Hospital, China
| | - Yanan Ma
- Medical Laboratory Department, Tianjin Third Central Hospital, China
| | - Lei Zhang
- Medical Laboratory Department, Tianjin Third Central Hospital, China
| | - Ni Li
- Obstetrics and Gynecology Program, Tianjin Third Central Hospital, China
| | - Jiandong Zhang
- Medical Laboratory Department, Tianjin Third Central Hospital, China
| |
Collapse
|
27
|
Mönckeberg M, Arias V, Fuenzalida R, Álvarez S, Toro V, Calvo A, Kusanovic JP, Monteiro LJ, Schepeler M, Nien JK, Martinez J, Illanes SE. Diagnostic Performance of First Trimester Screening of Preeclampsia Based on Uterine Artery Pulsatility Index and Maternal Risk Factors in Routine Clinical Use. Diagnostics (Basel) 2020; 10:E182. [PMID: 32225087 PMCID: PMC7235780 DOI: 10.3390/diagnostics10040182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/26/2020] [Accepted: 03/04/2020] [Indexed: 01/18/2023] Open
Abstract
Preeclampsia is a pregnancy-specific disorder defined by new onset of hypertension and proteinuria after 20 weeks of gestation. The early detection of patients at risk of developing preeclampsia is crucial, however, predictive models are still controversial. We aim to evaluate the diagnostic performance of a predictive algorithm in the first trimester of pregnancy, in order to identify patients that will subsequently develop preeclampsia, and to study the effect of aspirin on reducing the rate of this complication in patients classified as high risk by this algorithm. A retrospective cohort including 1132 patients attending prenatal care at Clínica Dávila in Santiago, Chile, was conceived. The risk of developing preeclampsia (early and late onset) was calculated using algorithms previously described by Plasencia et al. Patients classified as high risk, in the first trimester of pregnancy, by these algorithms, were candidates to receive 100 mg/daily aspirin as prophylaxis at the discretion of the attending physician. The overall incidence of preeclampsia in this cohort was 3.5% (40/1132), and the model for early onset preeclampsia prediction detected 33% of patients with early onset preeclampsia. Among the 105 patients considered at high risk of developing preeclampsia, 56 received aspirin and 49 patients did not. Among those who received aspirin, 12% (7/56) developed preeclampsia, which is equal to the rate of preeclampsia (12% (6/49)) of those who did not receive this medication. Therefore, the diagnostic performance of an algorithm combining uterine artery Doppler and maternal factors in the first trimester predicted only one third of patients that developed preeclampsia. Among those considered at high risk for developing the disease using this algorithm, aspirin did not change the incidence of preeclampsia, however, this could be due either to the small study sample size or the type of the study, a retrospective, non-interventional cohort study.
Collapse
Affiliation(s)
- Max Mönckeberg
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universidad de Los Andes, Santiago 7620001, Chile; (M.M.); (M.S.); (J.K.N.)
| | - Valentina Arias
- Faculty of Medicine, Universidad de Los Andes, Santiago 7620001, Chile; (V.A.); (R.F.); (V.T.); (A.C.)
| | - Rosario Fuenzalida
- Faculty of Medicine, Universidad de Los Andes, Santiago 7620001, Chile; (V.A.); (R.F.); (V.T.); (A.C.)
| | - Santiago Álvarez
- Faculty of Medicine, Universidad de Los Andes, Santiago 7620001, Chile; (V.A.); (R.F.); (V.T.); (A.C.)
| | - Victoria Toro
- Faculty of Medicine, Universidad de Los Andes, Santiago 7620001, Chile; (V.A.); (R.F.); (V.T.); (A.C.)
| | - Andrés Calvo
- Faculty of Medicine, Universidad de Los Andes, Santiago 7620001, Chile; (V.A.); (R.F.); (V.T.); (A.C.)
| | - Juan P. Kusanovic
- Center for Research and Innovation in Maternal-Fetal Medicine (CIMAF), Hospital Sótero del Río, Santiago 8207257, Chile;
- Division of Obstetrics and Gynecology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Lara J. Monteiro
- Centre for Biomedical Research, Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de Los Andes, Santiago 7620001, Chile;
| | - Manuel Schepeler
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universidad de Los Andes, Santiago 7620001, Chile; (M.M.); (M.S.); (J.K.N.)
- Department of Obstetrics and Gynecology, Clínica Dávila, Santiago 8420384, Chile;
| | - Jyh K. Nien
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universidad de Los Andes, Santiago 7620001, Chile; (M.M.); (M.S.); (J.K.N.)
- Department of Obstetrics and Gynecology, Clínica Dávila, Santiago 8420384, Chile;
| | - Jaime Martinez
- Department of Obstetrics and Gynecology, Clínica Dávila, Santiago 8420384, Chile;
| | - Sebastián E. Illanes
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universidad de Los Andes, Santiago 7620001, Chile; (M.M.); (M.S.); (J.K.N.)
- Centre for Biomedical Research, Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de Los Andes, Santiago 7620001, Chile;
- Department of Obstetrics and Gynecology, Clínica Dávila, Santiago 8420384, Chile;
| |
Collapse
|
28
|
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
|
29
|
Lee SM, Lee EM, Park JK, Jeon HS, Oh S, Hong S, Jung YM, Kim BJ, Kim SM, Norwitz ER, Lee EB, Louangsenlath S, Park CW, Jun JK, Park JS, Lee DY. Metabolic Biomarkers In Midtrimester Maternal Plasma Can Accurately Predict Adverse Pregnancy Outcome in Patients with SLE. Sci Rep 2019; 9:15169. [PMID: 31645572 PMCID: PMC6811572 DOI: 10.1038/s41598-019-51285-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 09/16/2019] [Indexed: 12/13/2022] Open
Abstract
Patients with systemic lupus erythematosus (SLE) are at increased risk for adverse pregnancy outcome (APO). Accurate prediction of APO is critical to identify, counsel, and manage these high-risk patients. We undertook this study to identify novel biomarkers in mid-trimester maternal plasma to identify pregnant patients with SLE at increased risk of APOs. The study population consisted of pregnant women whose plasma was taken in mid-trimester and available for metabolic signature: (1) SLE and normal pregnancy outcome (Group 1, n = 21); (2) SLE with APO (Group 2, n = 12); and (3) healthy pregnant controls (Group 3, n = 10). Mid-trimester maternal plasma was analyzed for integrative profiles of primary metabolite and phospholipid using gas chromatography time-of-flight mass spectrometry (GC-TOF MS) and liquid chromatography Orbitrap mass spectrometry (LC-Orbitrap MS). For performance comparison and validation, plasma samples were analyzed for sFlt-1/PlGF ratio. In the study population, APO developed in 12 of 33 women with SLE (36%). Metabolite profiling of mid-trimester maternal plasma samples identified a total of 327 metabolites using GC-TOF MS and LC-Orbitrap MS. Partial least squares discriminant analysis (PLS-DA) showed clear discrimination among the profiles of SLE groups and healthy pregnant controls (Groups 1/2 vs. 3). Moreover, direct comparison between Groups 1 and 2 demonstrated that 4 primary metabolites and 13 lipid molecules were significantly different. Binary logistic regression analysis suggested a potential metabolic biomarker model that could discriminate Groups 1 and 2. Receiver operating characteristic (ROC) analysis revealed the best predictability for APO with the combination model of two metabolites (LysoPC C22:5 and tryptophan) with AUC of 0.944, comparable to the AUC of sFlt-1/PlGF (AUC 0.857). In conclusion, metabolic biomarkers in mid-trimester maternal plasma can accurately predict APO in patients with SLE.
Collapse
Affiliation(s)
- Seung Mi Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Eun Mi Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute for Agricultural and Life Sciences, Seoul National University, Seoul, Korea
| | - Jin Kyun Park
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Hae Sun Jeon
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Sohee Oh
- Department of Biostatistics, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
| | - Subeen Hong
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Young Mi Jung
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Byoung Jae Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea.,Department of Obstetrics and Gynecology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
| | - Sun Min Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea.,Department of Obstetrics and Gynecology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
| | - Errol R Norwitz
- Department of Obstetrics and Gynecology, Tufts University School of Medicine, Boston, MA, USA
| | - Eun Bong Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | | | - Chan-Wook Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Jong Kwan Jun
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Joong Shin Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea.
| | - Do Yup Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute for Agricultural and Life Sciences, Seoul National University, Seoul, Korea.
| |
Collapse
|
30
|
Turkoglu O, Citil A, Katar C, Mert I, Kumar P, Yilmaz A, Uygur DS, Erkaya S, Graham SF, Bahado-Singh RO. Metabolomic identification of novel diagnostic biomarkers in ectopic pregnancy. Metabolomics 2019; 15:143. [PMID: 31630278 DOI: 10.1007/s11306-019-1607-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 10/11/2019] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Ectopic pregnancy (EP) is a potentially life-threatening condition and early diagnosis still remains a challenge, causing a delay in management leading to tubal rupture. OBJECTIVES To identify putative plasma biomarkers for the detection of tubal EP and elucidate altered biochemical pathways in EP compared to intrauterine pregnancies. METHODS This case-control study included prospective recruitment of 39 tubal EP cases and 89 early intrauterine pregnancy controls. Plasma samples were biochemically profiled using proton nuclear magnetic resonance spectroscopy (1H NMR). To avoid over-fitting, datasets were randomly divided into a discovery group (26 cases vs 60 controls) and a test group (13 cases and 29 controls). Logistic regression models were developed in the discovery group and validated in the independent test group. Area under the receiver operating characteristics curve (AUC), 95% confidence interval (CI), sensitivity, and specificity values were calculated. RESULTS In total 13 of 43 (30.3%) metabolite concentrations were significantly altered in EP plasma (p < 0.05). Metabolomic profiling yielded significant separation between EP and controls (p < 0.05). Independent validation of a two-metabolite model consisting of lactate and acetate, achieved an AUC (95% CI) = 0.935 (0.843-1.000) with a sensitivity of 92.3% and specificity of 96.6%. The second metabolite model (D-glucose, pyruvate, acetoacetate) performed well with an AUC (95% CI) = 0.822 (0.657-0.988) and a sensitivity of 84.6% and specificity of 86.2%. CONCLUSION We report novel metabolomic biomarkers with a high accuracy for the detection of EP. Accurate biomarkers could potentially result in improved early diagnosis of tubal EP cases.
Collapse
Affiliation(s)
- Onur Turkoglu
- Department of Obstetrics and Gynecology, Beaumont Health System, Royal Oak, MI, USA.
- Oakland University-William Beaumont School of Medicine, Rochester, MI, USA.
| | - Ayse Citil
- Department of Obstetrics and Gynecology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey
| | - Ceren Katar
- Department of Obstetrics and Gynecology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey
| | - Ismail Mert
- Division of Gynecological Oncology, Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA
| | - Praveen Kumar
- Department of Obstetrics and Gynecology, Beaumont Health System, Royal Oak, MI, USA
| | - Ali Yilmaz
- Department of Obstetrics and Gynecology, Beaumont Health System, Royal Oak, MI, USA
| | - Dilek S Uygur
- Department of Obstetrics and Gynecology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey
| | - Salim Erkaya
- Department of Obstetrics and Gynecology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey
| | - Stewart F Graham
- Department of Obstetrics and Gynecology, Beaumont Health System, Royal Oak, MI, USA
- Oakland University-William Beaumont School of Medicine, Rochester, MI, USA
| | - Ray O Bahado-Singh
- Department of Obstetrics and Gynecology, Beaumont Health System, Royal Oak, MI, USA
- Oakland University-William Beaumont School of Medicine, Rochester, MI, USA
| |
Collapse
|
31
|
Vonck S, Staelens AS, Lanssens D, Tomsin K, Oben J, Bruckers L, Gyselaers W. Development of a biophysical screening model for gestational hypertensive diseases. J Biomed Sci 2019; 26:38. [PMID: 31109316 PMCID: PMC6528347 DOI: 10.1186/s12929-019-0530-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 05/05/2019] [Indexed: 02/08/2023] Open
Abstract
Background To investigate the possibility of using maternal biophysical parameters only in screening for the different types of gestational hypertensive diseases. Methods A total of 969 pregnant women were randomly screened in first and second trimester, of which 8 developed Early-onset Preeclampsia, 29 Late-onset Preeclampsia, 35 Gestational Hypertension and 897 women had a normal outcome. An observational maternal hemodynamics assessment was done via standardized electrocardiogram-Doppler ultrasonography, Impedance Cardiography and bio-impedance, acquiring functional information on heart, arteries, veins and body fluid. Preliminary prediction models were developed to test the screening potential for early preeclampsia, late preeclampsia and gestational hypertension using a Partial Least Square Discriminant Analysis. Results A combined model using maternal characteristics with cardiovascular parameters in first and second trimester offers high screening performance with Area Under the Curve of 99,9% for Early-onset Preeclampsia, 95,3% for Late-onset Preeclampsia and 94% for Gestational Hypertension. Conclusions Using biophysical parameters as fundament for a new prediction model, without the need of biochemical parameters, seems feasible. However, validation in a large prospective study will reveal its true potential.
Collapse
Affiliation(s)
- Sharona Vonck
- Faculty of Medicine and Life Sciences, Hasselt University, Agoralaan, 3590, Diepenbeek, Belgium. .,Department of Obstetrics & Gynaecology, Ziekenhuis Oost-Limburg, Schiepse Bos 6, 3600, Genk, Belgium.
| | - Anneleen S Staelens
- Department of Obstetrics & Gynaecology, Ziekenhuis Oost-Limburg, Schiepse Bos 6, 3600, Genk, Belgium
| | - Dorien Lanssens
- Faculty of Medicine and Life Sciences, Hasselt University, Agoralaan, 3590, Diepenbeek, Belgium.,Department of Obstetrics & Gynaecology, Ziekenhuis Oost-Limburg, Schiepse Bos 6, 3600, Genk, Belgium
| | - Kathleen Tomsin
- Department of Obstetrics & Gynaecology, Ziekenhuis Oost-Limburg, Schiepse Bos 6, 3600, Genk, Belgium
| | - Jolien Oben
- Department of Obstetrics & Gynaecology, Ziekenhuis Oost-Limburg, Schiepse Bos 6, 3600, Genk, Belgium
| | - Liesbeth Bruckers
- Interuniversity Institute for Biostatistics and statistical Bioinformatics, Hasselt University, Agoralaan, 3590, Diepenbeek, Belgium
| | - Wilfried Gyselaers
- Faculty of Medicine and Life Sciences, Hasselt University, Agoralaan, 3590, Diepenbeek, Belgium.,Department Physiology, Hasselt University, Agoralaan, 3590, Diepenbeek, Belgium
| |
Collapse
|
32
|
Orosz L, Orosz G, Veress L, Dosa D, Orosz L, Arany I, Fabian A, Medve L, Pap K, Karanyi Z, Toth Z, Poka R, Than NG, Torok O. Screening for preeclampsia in the first trimester of pregnancy in routine clinical practice in Hungary. J Biotechnol 2019; 300:11-19. [PMID: 31055145 DOI: 10.1016/j.jbiotec.2019.04.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/18/2019] [Accepted: 04/20/2019] [Indexed: 12/01/2022]
Abstract
We aimed to evaluate the contribution of different factors in the Fetal Medicine Foundation algorithms for preeclampsia (PE) risk calculation during first-trimester screening in Hungary. We selected subjects for the nested case-control study from a prospective cohort of 2545 low-risk pregnancies. Eighty-two patients with PE and 82 gestational age-matched controls were included. Individual PE risk was calculated using two risk-assessing softwares. Using Astraia 2.3.1, considering maternal characteristics and biophysical parameters only, detection rates (DR) were 63.6% for early-PE and 67.6% for late-PE. When we added placenta associated plasma protein A (PAPP-A) to the risk calculation, DRs decreased to 54.5% and 64.8% respectively. Using Astraia 2.8.2 with maternal characteristics and biophysical parameters resulted in the DRs of 63.6% (early-PE) and 56.3% (late-PE). If we added PAPP-A to the risk calculation, DRs improved to 72.7% and 54.9%. The addition of placental growth factor (PlGF) did not increase detection rates in either calculation. In conclusion, using maternal characteristics, biophysical parameters, and PAPP-A, an acceptable screening efficacy could be achieved for early-PE during first-trimester screening. Since PlGF did not improve efficacy in our study, we suggest setting new standard curves for PlGF in Eastern European pregnant women, and the evaluation of novel biochemical markers.
Collapse
Affiliation(s)
- Laszlo Orosz
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen Medical and Health Science Centre, Nagyerdei korut 98, 4032, Debrecen, Hungary
| | - Gergo Orosz
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen Medical and Health Science Centre, Nagyerdei korut 98, 4032, Debrecen, Hungary
| | - Lajos Veress
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen Medical and Health Science Centre, Nagyerdei korut 98, 4032, Debrecen, Hungary
| | - Diana Dosa
- Department of Family and Occupational Medicine, Faculty of Public Health and Faculty of Medicine, University of Debrecen Medical and Health Science Centre, Moricz Zsigmond krt. 22, 4032, Debrecen, Hungary
| | - Laszlo Orosz
- Departement of Obststrics and Gynaecology, Andras Josa County and Teaching Hospital, Szent Istvan ut. 68, 4400, Nyiregyhaza, Hungary.
| | - Ibolya Arany
- Departement of Neonatology, Andras Josa County and Teaching Hospital, Szent Istvan ut. 68, 4400, Nyiregyhaza, Hungary
| | - Antal Fabian
- Departement of Obststrics and Gynaecology, Andras Josa County and Teaching Hospital, Szent Istvan ut. 68, 4400, Nyiregyhaza, Hungary
| | - Laszlo Medve
- Departement of Obststrics and Gynaecology, Andras Josa County and Teaching Hospital, Szent Istvan ut. 68, 4400, Nyiregyhaza, Hungary
| | - Karoly Pap
- Departement of Obststrics and Gynaecology, Andras Josa County and Teaching Hospital, Szent Istvan ut. 68, 4400, Nyiregyhaza, Hungary
| | - Zsolt Karanyi
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen Medical and Health Science Centre, Nagyerdei korut 98, 4032, Debrecen, Hungary
| | - Zoltan Toth
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen Medical and Health Science Centre, Nagyerdei korut 98, 4032, Debrecen, Hungary
| | - Robert Poka
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen Medical and Health Science Centre, Nagyerdei korut 98, 4032, Debrecen, Hungary
| | - Nandor Gabor Than
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudosok krt. 2, 1117, Budapest, Hungary; Maternity Private Clinic of Obstetrics and Gynecology, Kiralyhago ter 8, 1126, Budapest, Hungary; First Department of Pathology and Experimental Cancer Research, Semmelweis University, Ulloi ut 26, 1085, Budapest, Hungary.
| | - Olga Torok
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen Medical and Health Science Centre, Nagyerdei korut 98, 4032, Debrecen, Hungary.
| |
Collapse
|
33
|
Affiliation(s)
- Sarosh Rana
- From the Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago, IL (S.R.)
| | - Elizabeth Lemoine
- Harvard Medical School, Boston, MA (E.L.)
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (E.L., S.A.K.)
| | - Joey P. Granger
- Department of Physiology, University of Mississippi Medical Center, Jackson (J.P.G.)
| | - S. Ananth Karumanchi
- Departments of Medicine, Obstetrics and Gynecology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (S.A.K.)
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (E.L., S.A.K.)
| |
Collapse
|
34
|
Souza RT, Mayrink J, Leite DF, Costa ML, Calderon IM, Rocha EA, Vettorazzi J, Feitosa FE, Cecatti JG, Preterm SAMBA Study Group. Metabolomics applied to maternal and perinatal health: a review of new frontiers with a translation potential. Clinics (Sao Paulo) 2019; 74:e894. [PMID: 30916173 PMCID: PMC6438130 DOI: 10.6061/clinics/2019/e894] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/27/2018] [Indexed: 12/31/2022] Open
Abstract
The prediction or early diagnosis of maternal complications is challenging mostly because the main conditions, such as preeclampsia, preterm birth, fetal growth restriction, and gestational diabetes mellitus, are complex syndromes with multiple underlying mechanisms related to their occurrence. Limited advances in maternal and perinatal health in recent decades with respect to preventing these disorders have led to new approaches, and "omics" sciences have emerged as a potential field to be explored. Metabolomics is the study of a set of metabolites in a given sample and can represent the metabolic functioning of a cell, tissue or organism. Metabolomics has some advantages over genomics, transcriptomics, and proteomics, as metabolites are the final result of the interactions of genes, RNAs and proteins. Considering the recent "boom" in metabolomic studies and their importance in the research agenda, we here review the topic, explaining the rationale and theory of the metabolomic approach in different areas of maternal and perinatal health research for clinical practitioners. We also demonstrate the main exploratory studies of these maternal complications, commenting on their promising findings. The potential translational application of metabolomic studies, especially for the identification of predictive biomarkers, is supported by the current findings, although they require external validation in larger datasets and with alternative methodologies.
Collapse
Affiliation(s)
- Renato Teixeira Souza
- Departamento de Ginecologia e Obstetricia, Faculdade de Ciencias Medicas, Universidade Estadual de Campinas, Campinas, SP, BR
| | - Jussara Mayrink
- Departamento de Ginecologia e Obstetricia, Faculdade de Ciencias Medicas, Universidade Estadual de Campinas, Campinas, SP, BR
| | - Débora Farias Leite
- Departamento de Ginecologia e Obstetricia, Faculdade de Ciencias Medicas, Universidade Estadual de Campinas, Campinas, SP, BR
- Departamento Materno Infantil, Faculdade de Medicina, Universidade Federal de Pernambuco, Pernambuco, PE, BR
| | - Maria Laura Costa
- Departamento de Ginecologia e Obstetricia, Faculdade de Ciencias Medicas, Universidade Estadual de Campinas, Campinas, SP, BR
| | - Iracema Mattos Calderon
- Departamento de Ginecologia e Obstetricia, Faculdade de Medicina de Botucatu, Universidade Estadual de Sao Paulo (UNESP), Botucatu, SP, BR
| | - Edilberto Alves Rocha
- Departamento Materno Infantil, Faculdade de Medicina, Universidade Federal de Pernambuco, Pernambuco, PE, BR
| | - Janete Vettorazzi
- Departamento de Ginecologia e Obstetricia, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, RS, BR
| | - Francisco Edson Feitosa
- Departamento de Ginecologia e Obstetricia, Faculdade de Medicina, Universidade Federal do Ceara, Ceara, CE, BR
| | - José Guilherme Cecatti
- Departamento de Ginecologia e Obstetricia, Faculdade de Ciencias Medicas, Universidade Estadual de Campinas, Campinas, SP, BR
- Corresponding author. E-mail:
| | | |
Collapse
|
35
|
Delplancke TDJ, Wu Y, Han TL, Joncer LR, Qi H, Tong C, Baker PN. Metabolomics of Pregnancy Complications: Emerging Application of Maternal Hair. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2815439. [PMID: 30662903 PMCID: PMC6312607 DOI: 10.1155/2018/2815439] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/18/2018] [Indexed: 02/01/2023]
Abstract
In recent years, the study of metabolomics has begun to receive increasing international attention, especially as it pertains to medical research. This is due in part to the potential for discovery of new biomarkers in the metabolome and to a new understanding of the "exposome", which refers to the endogenous and exogenous compounds that reflect external exposures. Consequently, metabolomics research into pregnancy-related issues has increased. Biomarkers discovered through metabolomics may shed some light on the etiology of certain pregnancy-related complications and their adverse effects on future maternal health and infant development and improve current clinical management. The discoveries and methods used in these studies will be compiled and summarized within the following paper. A further focus of this paper is the use of hair as a biological sample, which is gaining increasing attention across diverse fields due to its noninvasive sampling method and the metabolome stability. Its significance in exposome studies will be considered in this review, as well as the potential to associate exposures with adverse pregnancy outcomes. Currently, hair has been used in only two metabolomics studies relating to fetal growth restriction (FGR) and gestational diabetes mellitus (GDM).
Collapse
Affiliation(s)
- Thibaut D. J. Delplancke
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing Medical University, Chongqing 400016, China
- International Collaborative Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Yue Wu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing Medical University, Chongqing 400016, China
- International Collaborative Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Ting-Li Han
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing Medical University, Chongqing 400016, China
- International Collaborative Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Lingga R. Joncer
- International Collaborative Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Hongbo Qi
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing Medical University, Chongqing 400016, China
- International Collaborative Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Chao Tong
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing Medical University, Chongqing 400016, China
- International Collaborative Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Philip N. Baker
- International Collaborative Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
- Liggins Institute, University of Auckland, Auckland, New Zealand
- College of Medicine, University of Leicester, Leicester LE1 7RH, UK
| |
Collapse
|
36
|
Mayrink J, Costa ML, Cecatti JG. Preeclampsia in 2018: Revisiting Concepts, Physiopathology, and Prediction. ScientificWorldJournal 2018; 2018:6268276. [PMID: 30622442 PMCID: PMC6304478 DOI: 10.1155/2018/6268276] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/05/2018] [Accepted: 11/22/2018] [Indexed: 12/13/2022] Open
Abstract
Preeclampsia currently remains one of the leading causes of death and severe maternal morbidity. Although its prevalence is still underestimated in some places due to underreporting, preeclampsia is a disease that health professionals need to know how to deal with and take action. For this reason, the studies about the theme remain along with the advances in their understanding that often implies improvement and change of concepts and conducts. The complexity of its etiology is a challenge and requires further studies for its full understanding. Apparently, poor adaptation of the maternal organism to the conceptus, marked by the nonoccurrence of changes in the uterine spiral arteries, determines a series of systemic repercussions that compound the various forms of preeclampsia presentation. In recent years, the use of acetylsalicylic acid to prevent cases of early onset of the disease has been consolidated and, alongside, studies have advanced the development of accessible and effective methods of identifying women at risk of preeclampsia. The aim of this review is to discuss updates on the occurrence, concept, pathophysiology, repercussion, prevention, and prediction of preeclampsia.
Collapse
Affiliation(s)
- J. Mayrink
- Obstetric Unit, Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - M. L. Costa
- Obstetric Unit, Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - J. G. Cecatti
- Obstetric Unit, Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas, Campinas, Brazil
| |
Collapse
|
37
|
Jääskeläinen T, Kärkkäinen O, Jokkala J, Litonius K, Heinonen S, Auriola S, Lehtonen M, Hanhineva K, Laivuori H. A Non-Targeted LC-MS Profiling Reveals Elevated Levels of Carnitine Precursors and Trimethylated Compounds in the Cord Plasma of Pre-Eclamptic Infants. Sci Rep 2018; 8:14616. [PMID: 30279541 PMCID: PMC6168522 DOI: 10.1038/s41598-018-32804-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 09/14/2018] [Indexed: 11/21/2022] Open
Abstract
Preeclampsia (PE) is a complex pregnancy disorder. It is not extensively known how the metabolic alterations of PE women contribute to the metabolism of newborn. We applied liquid chromatography-mass spectrometry (LC-MS) based non-targeted metabolomics to determine whether the metabolic profile of plasma from umbilical cord differs between infants born to PE and non-PE pregnancies in the FINNPEC study. Cord plasma was available from 42 newborns born from PE and 53 from non-PE pregnancies. 133 molecular features differed between PE and non-PE newborns after correction for multiple testing. Decreased levels of 4-pyridoxic acid were observed in the cord plasma samples of PE newborns when compared to non-PE newborns. Compounds representing following areas of metabolism were increased in the cord plasma of PE newborns: urea and creatine metabolism; carnitine biosynthesis and acylcarnitines; putrescine metabolites; tryptophan metabolism and phosphatidylcholines. To our knowledge, this study is the first one to apply LC-MS based metabolomics in cord plasma of PE newborns. We demonstrate that this strategy provides a global picture of the widespread metabolic alterations associated with PE and particularly the elevated levels of carnitine precursors and trimethylated compounds appear to be associated with PE at birth.
Collapse
Affiliation(s)
- Tiina Jääskeläinen
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
| | - Olli Kärkkäinen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Jenna Jokkala
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Kaisa Litonius
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Seppo Heinonen
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Marko Lehtonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Kati Hanhineva
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Hannele Laivuori
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital and University of Tampere, Faculty of Medicine and Life Sciences, Tampere, Finland
| |
Collapse
|
38
|
Miranda J, Simões RV, Paules C, Cañueto D, Pardo-Cea MA, García-Martín ML, Crovetto F, Fuertes-Martin R, Domenech M, Gómez-Roig MD, Eixarch E, Estruch R, Hansson SR, Amigó N, Cañellas N, Crispi F, Gratacós E. Metabolic profiling and targeted lipidomics reveals a disturbed lipid profile in mothers and fetuses with intrauterine growth restriction. Sci Rep 2018; 8:13614. [PMID: 30206284 PMCID: PMC6134091 DOI: 10.1038/s41598-018-31832-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/13/2018] [Indexed: 12/11/2022] Open
Abstract
Fetal growth may be impaired by poor placental function or maternal conditions, each of which can influence the transfer of nutrients and oxygen from the mother to the developing fetus. Large-scale studies of metabolites (metabolomics) are key to understand cellular metabolism and pathophysiology of human conditions. Herein, maternal and cord blood plasma samples were used for NMR-based metabolic fingerprinting and profiling, including analysis of the enrichment of circulating lipid classes and subclasses, as well as the number of sub-fraction particles and their size. Changes in phosphatidylcholines and glycoproteins were prominent in growth-restricted fetuses indicating significant alterations in their abundance and biophysical properties. Lipoprotein profiles showed significantly lower plasma concentrations of cholesterol-intermediate density lipoprotein (IDL), triglycerides-IDL and high-density lipoprotein (HDL) in mothers of growth-restricted fetuses compared to controls (p < 0.05). In contrast, growth-restricted fetuses had significantly higher plasma concentrations of cholesterol and triglycerides transporting lipoproteins [LDL, IDL, and VLDL, (p < 0.005; all)], as well as increased VLDL particle types (large, medium and small). Significant changes in plasma concentrations of formate, histidine, isoleucine and citrate in growth-restricted fetuses were also observed. Comprehensive metabolic profiling reveals that both, mother and fetuses of pregnancies complicated with fetal growth restriction have a substantial disruption in lipid metabolism.
Collapse
Affiliation(s)
- Jezid Miranda
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Rui V Simões
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Cristina Paules
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Daniel Cañueto
- Metabolomics Platform, IISPV, DEEiA, Universidad Rovira i Virgili, Tarragona, Spain
| | | | - María L García-Martín
- BIONAND, Andalusian Centre for Nanomedicine and Biotechnology, Junta de Andalucía, Universidad de Málaga, Málaga, Spain
| | - Francesca Crovetto
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Rocio Fuertes-Martin
- Metabolomics Platform, IISPV, DEEiA, Universidad Rovira i Virgili, Tarragona, Spain
- Biosfer Teslab, Reus, Spain
| | - Monica Domenech
- Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - María D Gómez-Roig
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Elisenda Eixarch
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Ramon Estruch
- Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Stefan R Hansson
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Lund University, Sweden; Skåne University Hospital, Lund, Sweden
| | | | - Nicolau Cañellas
- Metabolomics Platform, IISPV, DEEiA, Universidad Rovira i Virgili, Tarragona, Spain
- CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Madrid, Spain
| | - Fatima Crispi
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain.
| | - Eduard Gratacós
- Fetal i+D Fetal Medicine Research, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| |
Collapse
|
39
|
Abstract
Preeclampsia is a multifactorial disorder defined by hypertension and increased urinary protein excretion during pregnancy. It is a significant cause of maternal and neonatal deaths worldwide. Despite various research efforts to clarify pathogenies of preeclampsia and predict this disease before beginning of symptoms, the pathogenesis of preeclampsia is unclear. Early prediction and diagnosis of women at risk of preeclampsia has not markedly improved. Therefore, the objective of this study was to perform a review on metabolomic articles assessing predictive and diagnostic biomarkers of preeclampsia. Four electronic databases including PubMed/Medline, Web of Science, Sciencedirect, and Scopus were searched to identify studies of preeclampsia in humans using metabolomics from inception to March 2018. Twenty-one articles in a variety of biological specimens and analytical platforms were included in the present review. Metabolite profiles may assist in the diagnosis of preeclampsia and discrimination of its subtypes. Lipids and their related metabolites were the most generally detected metabolites. Although metabolomic biomarkers of preeclampsia are not routinely used, this review suggests that metabolomics has the potential to be developed into a clinical tool for preeclampsia diagnosis and could contribute to an improved understanding of disease mechanisms. ABBREVIATIONS PE: preeclampsia; sFlt-1: soluble FMS-like tyrosine kinase-1; PlGF: placental growth factor; GC-MS: gas chromatography-mass spectrometry; LC-MS: liquid chromatography-mass spectrometry; NMR: nuclear magnetic resonance spectroscopy; HMDB: human metabolome database; RCT: randomized control trial; e-PE: early-onset PE; l-PE: late-onset PE; PLS-DA: partial least-squares-discriminant analysis; CRL: crown-rump length; UtPI: uterine artery Doppler pulsatility index; BMI: body mass index; MAP: mean arterial pressure; OS: oxidative stress; PAPPA: plasma protein A; FTIR: Fourier transform infrared; BCAA: branched chain amino acids; Arg: arginine; NO: nitric oxide.
Collapse
Affiliation(s)
- B Fatemeh Nobakht M Gh
- a Department of Basic Medical Sciences , Neyshabur University of Medical Sciences , Neyshabur , Iran
| |
Collapse
|
40
|
Tejera E, Cruz-Monteagudo M, Burgos G, Sánchez ME, Sánchez-Rodríguez A, Pérez-Castillo Y, Borges F, Cordeiro MNDS, Paz-Y-Miño C, Rebelo I. Consensus strategy in genes prioritization and combined bioinformatics analysis for preeclampsia pathogenesis. BMC Med Genomics 2017; 10:50. [PMID: 28789679 PMCID: PMC5549357 DOI: 10.1186/s12920-017-0286-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/28/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Preeclampsia is a multifactorial disease with unknown pathogenesis. Even when recent studies explored this disease using several bioinformatics tools, the main objective was not directed to pathogenesis. Additionally, consensus prioritization was proved to be highly efficient in the recognition of genes-disease association. However, not information is available about the consensus ability to early recognize genes directly involved in pathogenesis. Therefore our aim in this study is to apply several theoretical approaches to explore preeclampsia; specifically those genes directly involved in the pathogenesis. METHODS We firstly evaluated the consensus between 12 prioritization strategies to early recognize pathogenic genes related to preeclampsia. A communality analysis in the protein-protein interaction network of previously selected genes was done including further enrichment analysis. The enrichment analysis includes metabolic pathways as well as gene ontology. Microarray data was also collected and used in order to confirm our results or as a strategy to weight the previously enriched pathways. RESULTS The consensus prioritized gene list was rationally filtered to 476 genes using several criteria. The communality analysis showed an enrichment of communities connected with VEGF-signaling pathway. This pathway is also enriched considering the microarray data. Our result point to VEGF, FLT1 and KDR as relevant pathogenic genes, as well as those connected with NO metabolism. CONCLUSION Our results revealed that consensus strategy improve the detection and initial enrichment of pathogenic genes, at least in preeclampsia condition. Moreover the combination of the first percent of the prioritized genes with protein-protein interaction network followed by communality analysis reduces the gene space. This approach actually identifies well known genes related with pathogenesis. However, genes like HSP90, PAK2, CD247 and others included in the first 1% of the prioritized list need to be further explored in preeclampsia pathogenesis through experimental approaches.
Collapse
Affiliation(s)
- Eduardo Tejera
- Facultad de Medicina, Universidad de Las Américas, Av. de los Granados E12-41y Colimes esq, EC170125, Quito, Ecuador.
| | - Maykel Cruz-Monteagudo
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine and Center for Computational Science, University of Miami, FL 33136, Miami, USA.,Department of General Education, West Coast University-Miami Campus, Doral, FL 33178, USA.,CIQUP/Departamento de Quimica e Bioquimica, Faculdade de Ciências, Universidade do Porto, 4169-007, Porto, Portugal.,REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007, Porto, Portugal
| | - Germán Burgos
- Facultad de Medicina, Universidad de Las Américas, Av. de los Granados E12-41y Colimes esq, EC170125, Quito, Ecuador
| | - María-Eugenia Sánchez
- Facultad de Medicina, Universidad de Las Américas, Av. de los Granados E12-41y Colimes esq, EC170125, Quito, Ecuador
| | - Aminael Sánchez-Rodríguez
- Departamento de Ciencias Naturales, Universidad Técnica Particular de Loja, Calle París S/N, EC1101608, Loja, Ecuador
| | | | - Fernanda Borges
- CIQUP/Departamento de Quimica e Bioquimica, Faculdade de Ciências, Universidade do Porto, 4169-007, Porto, Portugal
| | | | - César Paz-Y-Miño
- Centro de Investigaciones genética y genómica, Facultad de Ciencias de la Salud, Universidad Tecnológica Equinoccial, Quito, Ecuador
| | - Irene Rebelo
- Faculty of Pharmacy, University of Porto, Porto, Portugal.,UCIBIO@REQUIMTE, Caparica, Portugal
| |
Collapse
|
41
|
Romero R, Erez O, Maymon E, Chaemsaithong P, Xu Z, Pacora P, Chaiworapongsa T, Done B, Hassan SS, Tarca AL. The maternal plasma proteome changes as a function of gestational age in normal pregnancy: a longitudinal study. Am J Obstet Gynecol 2017; 217:67.e1-67.e21. [PMID: 28263753 PMCID: PMC5813489 DOI: 10.1016/j.ajog.2017.02.037] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/10/2017] [Accepted: 02/23/2017] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Pregnancy is accompanied by dramatic physiological changes in maternal plasma proteins. Characterization of the maternal plasma proteome in normal pregnancy is an essential step for understanding changes to predict pregnancy outcome. The objective of this study was to describe maternal plasma proteins that change in abundance with advancing gestational age and determine biological processes that are perturbed in normal pregnancy. STUDY DESIGN A longitudinal study included 43 normal pregnancies that had a term delivery of an infant who was appropriate for gestational age without maternal or neonatal complications. For each pregnancy, 3 to 6 maternal plasma samples (median, 5) were profiled to measure the abundance of 1125 proteins using multiplex assays. Linear mixed-effects models with polynomial splines were used to model protein abundance as a function of gestational age, and the significance of the association was inferred via likelihood ratio tests. Proteins considered to be significantly changed were defined as having the following: (1) >1.5-fold change between 8 and 40 weeks of gestation; and (2) a false discovery rate-adjusted value of P < .1. Gene ontology enrichment analysis was used to identify biological processes overrepresented among the proteins that changed with advancing gestation. RESULTS The following results were found: (1) Ten percent (112 of 1125) of the profiled proteins changed in abundance as a function of gestational age; (2) of the 1125 proteins analyzed, glypican-3, sialic acid-binding immunoglobulin-type lectin-6, placental growth factor, C-C motif-28, carbonic anhydrase 6, prolactin, interleukin-1 receptor 4, dual-specificity mitogen-activated protein kinase 4, and pregnancy-associated plasma protein-A had more than a 5-fold change in abundance across gestation (these 9 proteins are known to be involved in a wide range of both physiological and pathological processes, such as growth regulation, embryogenesis, angiogenesis immunoregulation, inflammation etc); and (3) biological processes associated with protein changes in normal pregnancy included defense response, defense response to bacteria, proteolysis, and leukocyte migration (false discovery rate, 10%). CONCLUSION The plasma proteome of normal pregnancy demonstrates dramatic changes in both the magnitude of changes and the fraction of the proteins involved. Such information is important to understand the physiology of pregnancy and the development of biomarkers to differentiate normal vs abnormal pregnancy and determine the response to interventions.
Collapse
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI.
| | - Offer Erez
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Eli Maymon
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Piya Chaemsaithong
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Zhonghui Xu
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI
| | - Percy Pacora
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Bogdan Done
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI
| | - Sonia S Hassan
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Adi L Tarca
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI.
| |
Collapse
|
42
|
Kelly RS, Giorgio RT, Chawes BL, Palacios NI, Gray KJ, Mirzakhani H, Wu A, Blighe K, Weiss ST, Lasky-Su J. Applications of Metabolomics in the Study and Management of Preeclampsia; A Review of the Literature. Metabolomics 2017; 13:86. [PMID: 30473646 PMCID: PMC6247796 DOI: 10.1007/s11306-017-1225-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 06/05/2017] [Indexed: 12/12/2022]
Abstract
Introduction Preeclampsia represents a major public health burden worldwide, but predictive and diagnostic biomarkers are lacking. Metabolomics is emerging as a valuable approach to generating novel biomarkers whilst increasing the mechanistic understanding of this complex condition. Objectives To summarize the published literature on the use of metabolomics as a tool to study preeclampsia. Methods PubMed and Web of Science were searched for articles that performed metabolomic profiling of human biosamples using either Mass-spectrometry or Nuclear Magnetic Resonance based approaches and which included preeclampsia as a primary endpoint. Results Twenty-eight studies investigating the metabolome of preeclampsia in a variety of biospecimens were identified. Individual metabolite and metabolite profiles were reported to have discriminatory ability to distinguish preeclamptic from normal pregnancies, both prior to and post diagnosis. Lipids and carnitines were among the most commonly reported metabolites. Further work and validation studies are required to demonstrate the utility of such metabolites as preeclampsia biomarkers. Conclusion Metabolomic-based biomarkers of preeclampsia have yet to be integrated into routine clinical practice. However, metabolomic profiling is becoming increasingly popular in the study of preeclampsia and is likely to be a valuable tool to better understand the pathophysiology of this disorder and to better classify its subtypes, particularly when integrated with other omic data.
Collapse
Affiliation(s)
- Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital Harvard Medical School, Boston MA 02115, USA
| | - Rachel T Giorgio
- Channing Division of Network Medicine, Brigham and Women's Hospital Harvard Medical School, Boston MA 02115, USA
| | - Bo L Chawes
- Channing Division of Network Medicine, Brigham and Women's Hospital Harvard Medical School, Boston MA 02115, USA
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Natalia I Palacios
- Department of Public Health University of Massachusetts, Lowell, Lowell MA
- Department of Nutrition, Harvard School of Public Health, Boston MA
| | - Kathryn J Gray
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Hoooman Mirzakhani
- Channing Division of Network Medicine, Brigham and Women's Hospital Harvard Medical School, Boston MA 02115, USA
| | - Ann Wu
- Boston Children's Hospital
| | - Kevin Blighe
- Channing Division of Network Medicine, Brigham and Women's Hospital Harvard Medical School, Boston MA 02115, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital Harvard Medical School, Boston MA 02115, USA
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital Harvard Medical School, Boston MA 02115, USA
| |
Collapse
|
43
|
Maier M, Brückmann A, Schleußner E, Schlembach D. Using critical flicker frequency in the evaluation of visual impairment in preeclamptic women. Eur J Obstet Gynecol Reprod Biol 2017; 211:188-193. [PMID: 28292692 DOI: 10.1016/j.ejogrb.2017.02.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 02/20/2017] [Accepted: 02/28/2017] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To assess critical flicker frequency (CFF) in normal uneventful pregnancy and preeclampsia. STUDY-DESIGN Case-control observational study at the University Hospital Jena and Outpatient Institute for Prenatal Diagnosis and Preventive Medicine. 25 non-pregnant women, 75 uncomplicated pregnant women in first, second and third trimester, and 15 women with overt preeclampsia. For comparison with preeclamptic patients we matched 15 normal pregnant women (mNP) for age, parity, body mass index, current smoking and family history of cardiovascular disease (CVD). We measured CFF using the portable HEPAtonorm Analyzer (nevoLAB GmbH, Germany). This device generates a flickering red light, starting with a frequency of 60Hz, giving the subjective an impression of a steady light. The participant signifies once the impression of a flickering light is recognized, and this CFF is recorded. Mean CFF and standard deviation is automatically calculated. Statistical analysis was performed using SPSS Version 22 for Windows. Following assessment of normal distribution with Kolmogorov-Smirnow test, comparisons were made with univariate and multivariate ANOVA and with unpaired and paired t test for continuous data and with χ2 test for categorical data. RESULTS Critical flicker frequency in healthy pregnant women does not differ from nonpregnant women. No significant differences in CFF measurements exist in first, second, and third trimester. In preeclampsia, CFF is significantly decreased compared to normal pregnant women (PE 38.80±2.16 vs. mNP 46.23±3.37; p=0.000). This alteration persists even some weeks postpartum (PE 41.17±1.13 vs. mNP 46.45±3.44; p=0.003). CONCLUSION In preeclamptic women, CFF is decreased indicating an altered endothelial situation. The finding that CFF remains reduced postpartum may be explained by either the effect of preeclampsia on maternal endothelium causing longer lasting damage or indicate a preexisting endothelial disorder. Up to this point, precise responsible mechanisms for altered CFF in preeclampsia are currently unclear and further studies are needed.
Collapse
Affiliation(s)
- Marina Maier
- Department of Obstetrics, Jena University Hospital, Friedrich-Schiller-University, 07744 Jena, Germany
| | - Andreas Brückmann
- Department of Prenatal Diagnosis and Preventive Medicine, GesaTal Medical Center, 99089 Erfurt, Germany
| | - Ekkehard Schleußner
- Department of Obstetrics, Jena University Hospital, Friedrich-Schiller-University, 07744 Jena, Germany
| | - Dietmar Schlembach
- Department of Obstetrics, Jena University Hospital, Friedrich-Schiller-University, 07744 Jena, Germany; Clinic of Obstetrics, Vivantes Hospital Berlin-Neukoelln, 12351 Berlin, Germany.
| |
Collapse
|
44
|
The role of aspirin dose on the prevention of preeclampsia and fetal growth restriction: systematic review and meta-analysis. Am J Obstet Gynecol 2017; 216:110-120.e6. [PMID: 27640943 DOI: 10.1016/j.ajog.2016.09.076] [Citation(s) in RCA: 426] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 08/25/2016] [Accepted: 09/07/2016] [Indexed: 11/22/2022]
Abstract
BACKGROUND Preeclampsia and fetal growth restriction are major causes of perinatal death and handicap in survivors. Randomized clinical trials have reported that the risk of preeclampsia, severe preeclampsia, and fetal growth restriction can be reduced by the prophylactic use of aspirin in high-risk women, but the appropriate dose of the drug to achieve this objective is not certain. OBJECTIVE We sought to estimate the impact of aspirin dosage on the prevention of preeclampsia, severe preeclampsia, and fetal growth restriction. STUDY DESIGN We performed a systematic review and meta-analysis of randomized controlled trials comparing the effect of daily aspirin or placebo (or no treatment) during pregnancy. We searched MEDLINE, Embase, Web of Science, and Cochrane Central Register of Controlled Trials up to December 2015, and study bibliographies were reviewed. Authors were contacted to obtain additional data when needed. Relative risks for preeclampsia, severe preeclampsia, and fetal growth restriction were calculated with 95% confidence intervals using random-effect models. Dose-response effect was evaluated using meta-regression and reported as adjusted R2. Analyses were stratified according to gestational age at initiation of aspirin (≤16 and >16 weeks) and repeated after exclusion of studies at high risk of biases. RESULTS In all, 45 randomized controlled trials included a total of 20,909 pregnant women randomized to between 50-150 mg of aspirin daily. When aspirin was initiated at ≤16 weeks, there was a significant reduction and a dose-response effect for the prevention of preeclampsia (relative risk, 0.57; 95% confidence interval, 0.43-0.75; P < .001; R2, 44%; P = .036), severe preeclampsia (relative risk, 0.47; 95% confidence interval, 0.26-0.83; P = .009; R2, 100%; P = .008), and fetal growth restriction (relative risk, 0.56; 95% confidence interval, 0.44-0.70; P < .001; R2, 100%; P = .044) with higher dosages of aspirin being associated with greater reduction of the 3 outcomes. Similar results were observed after the exclusion of studies at high risk of biases. When aspirin was initiated at >16 weeks, there was a smaller reduction of preeclampsia (relative risk, 0.81; 95% confidence interval, 0.66-0.99; P = .04) without relationship with aspirin dosage (R2, 0%; P = .941). Aspirin initiated at >16 weeks was not associated with a risk reduction or a dose-response effect for severe preeclampsia (relative risk, 0.85; 95% confidence interval, 0.64-1.14; P = .28; R2, 0%; P = .838) and fetal growth restriction (relative risk, 0.95; 95% confidence interval, 0.86-1.05; P = .34; R2, not available; P = .563). CONCLUSION Prevention of preeclampsia and fetal growth restriction using aspirin in early pregnancy is associated with a dose-response effect. Low-dose aspirin initiated at >16 weeks' gestation has a modest or no impact on the risk of preeclampsia, severe preeclampsia, and fetal growth restriction. Women at high risk for those outcomes should be identified in early pregnancy.
Collapse
|
45
|
Scazzocchio E, Crovetto F, Triunfo S, Gratacós E, Figueras F. Validation of a first-trimester screening model for pre-eclampsia in an unselected population. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2017; 49:188-193. [PMID: 27257033 DOI: 10.1002/uog.15982] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 04/26/2016] [Accepted: 05/27/2016] [Indexed: 05/07/2023]
Abstract
OBJECTIVE To validate the performance of a previously constructed first-trimester predictive model for pre-eclampsia (PE) in routine care of an unselected population. METHODS A validation cohort of 4621 consecutive women attending their routine first-trimester ultrasound examination was used to test a prediction model for PE that had been developed previously in 5170 women. The prediction model included maternal factors, uterine artery Doppler, blood pressure and pregnancy-associated plasma protein-A. Model performance was evaluated using receiver-operating characteristics (ROC) curve analysis and ROC curves from both cohorts were compared unpaired. RESULTS Among the 4203 women included in the final analysis, 169 (4.0%) developed PE, including 141 (3.4%) cases of late-onset PE and 28 (0.7%) cases of early-onset PE. For early-onset PE, the model showed an area under the ROC curve of 0.94 (95% CI, 0.88-0.99), which did not differ significantly (P = 0.37) from that obtained in the construction cohort (0.88 (95% CI, 0.78-0.99)). For late-onset PE, the final model showed an area under the ROC curve of 0.72 (95% CI, 0.66-0.77), which did not differ significantly (P = 0.49) from that obtained in the construction cohort (0.75 (95% CI, 0.67-0.82)). CONCLUSION The prediction model for PE achieved a similar performance to that obtained in the construction cohort when tested on a subsequent cohort of women, confirming its validity as a predictive model for PE. Copyright © 2016 ISUOG. Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- E Scazzocchio
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
- Obstetrics, Gynecology and Reproductive Medicine Department, Quirón Dexeus Universitari Hospital, Barcelona, Spain
| | - F Crovetto
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - S Triunfo
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - E Gratacós
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - F Figueras
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| |
Collapse
|
46
|
Kelly RS, Croteau-Chonka DC, Dahlin A, Mirzakhani H, Wu AC, Wan ES, McGeachie MJ, Qiu W, Sordillo JE, Al-Garawi A, Gray KJ, McElrath TF, Carey VJ, Clish CB, Litonjua AA, Weiss ST, Lasky-Su JA. Integration of metabolomic and transcriptomic networks in pregnant women reveals biological pathways and predictive signatures associated with preeclampsia. Metabolomics 2017; 13:7. [PMID: 28596717 PMCID: PMC5458629 DOI: 10.1007/s11306-016-1149-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Preeclampsia is a leading cause of maternal and fetal mortality worldwide, yet its exact pathogenesis remains elusive. OBJECTIVES This study, nested within the Vitamin D Antenatal Asthma Reduction Trial (VDAART), aimed to develop integrated omics models of preeclampsia that have utility in both prediction and in the elucidation of underlying biological mechanisms. METHODS Metabolomic profiling was performed on first trimester plasma samples of 47 pregnant women from VDAART who subsequently developed preeclampsia and 62 controls with healthy pregnancies, using liquid-chromatography tandem mass-spectrometry. Metabolomic profiles were generated based on logistic regression models and assessed using Received Operator Characteristic Curve analysis. These profiles were compared to profiles from generated using third trimester samples. The first trimester metabolite profile was then integrated with a pre-existing transcriptomic profile using network methods. RESULTS In total, 72 (0.9%) metabolite features were associated (p<0.01) with preeclampsia after adjustment for maternal age, race, and gestational age. These features had moderate to good discriminatory ability; in ROC curve analyses a summary score based on these features displayed an area under the curve (AUC) of 0.794 (95%CI 0.700, 0.888). This profile retained the ability to distinguish preeclamptic from healthy pregnancies in the third trimester (AUC:0.762 (95% CI 0.663, 0.860)). Additionally, metabolite set enrichment analysis identified common pathways, including glycerophospholipid metabolism, at the two time-points. Integration with the transcriptomic signature refined these results suggesting a particular role for lipid imbalance, immune function and the circulatory system. CONCLUSIONS These findings suggest it is possible to develop a predictive metabolomic profile of preeclampsia. This profile is characterized by changes in lipid and amino acid metabolism and dysregulation of immune response and can be refined through interaction with transcriptomic data. However validation in larger and more diverse populations is required.
Collapse
Affiliation(s)
- Rachel S. Kelly
- Channing Department of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Damien C. Croteau-Chonka
- Channing Department of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Amber Dahlin
- Channing Department of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Hooman Mirzakhani
- Channing Department of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ann C. Wu
- Channing Department of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA 02215, USA
| | - Emily S. Wan
- Channing Department of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Michael J. McGeachie
- Channing Department of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Weiliang Qiu
- Channing Department of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Joanne E. Sordillo
- Channing Department of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Amal Al-Garawi
- Channing Department of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Kathryn J. Gray
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Thomas F. McElrath
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Vincent J. Carey
- Channing Department of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Clary B. Clish
- Metabolomics Platform, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02141, USA
| | - Augusto A. Litonjua
- Channing Department of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Scott T. Weiss
- Channing Department of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jessica A. Lasky-Su
- Channing Department of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
47
|
Piras D, Locci E, Palmas F, Ferino G, Fanos V, Noto A, D’aloja E, Finco G. Rare disease: a focus on metabolomics. Expert Opin Orphan Drugs 2016. [DOI: 10.1080/21678707.2016.1252671] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
48
|
Bahado-Singh RO, Syngelaki A, Mandal R, Graham SF, Akolekar R, Han B, Bjondahl TC, Dong E, Bauer S, Alpay-Savasan Z, Turkoglu O, Ogunyemi D, Poon LC, Wishart DS, Nicolaides KH. Metabolomic determination of pathogenesis of late-onset preeclampsia. J Matern Fetal Neonatal Med 2016; 30:658-664. [PMID: 27569705 DOI: 10.1080/14767058.2016.1185411] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Our primary objective was to apply metabolomic pathway analysis of first trimester maternal serum to provide an insight into the pathogenesis of late-onset preeclampsia (late-PE) and thereby identify plausible therapeutic targets for PE. METHODS NMR-based metabolomics analysis was performed on 29 cases of late-PE and 55 unaffected controls. In order to achieve sufficient statistical power to perform the pathway analysis, these cases were combined with a group of previously analyzed specimens, 30 late-PE cases and 60 unaffected controls. Specimens from both groups of cases and controls were collected in the same clinical centers during the same time period. In addition, NMR analyses were performed in the same lab and using the same techniques. RESULTS We identified abnormalities in branch chain amino acids (valine, leucine and isoleucine) and propanoate, glycolysis, gluconeogenesis and ketone body metabolic pathways. The results suggest insulin resistance and metabolic syndrome, mitochondrial dysfunction and disturbance of energy metabolism, oxidative stress and lipid dysfunction in the pathogenesis of late PE and suggest a potential role for agents that reduce insulin resistance in PE. CONCLUSIONS Branched chain amino acids are known markers of insulin resistance and strongly predict future diabetes development. The analysis provides independent evidence linking insulin resistance and late-PE and suggests a potentially important therapeutic role for pharmacologic agents that reduce insulin resistance for late-PE.
Collapse
Affiliation(s)
- Ray O Bahado-Singh
- a Department of Obstetrics and Gynecology , Beaumont Health , Royal Oak , MI , USA
| | - Argyro Syngelaki
- b Harris Birthright Research Centre for Fetal Medicine , Division of Women's Health, King's College Hospital , London , UK
| | - Rupsari Mandal
- c Department of Biological Sciences , University of Alberta , Edmonton, Alberta , Canada , and
| | - Stewart F Graham
- a Department of Obstetrics and Gynecology , Beaumont Health , Royal Oak , MI , USA
| | - Ranjit Akolekar
- b Harris Birthright Research Centre for Fetal Medicine , Division of Women's Health, King's College Hospital , London , UK
| | - Beomsoo Han
- c Department of Biological Sciences , University of Alberta , Edmonton, Alberta , Canada , and
| | - Trent C Bjondahl
- c Department of Biological Sciences , University of Alberta , Edmonton, Alberta , Canada , and
| | - Edison Dong
- c Department of Biological Sciences , University of Alberta , Edmonton, Alberta , Canada , and
| | - Samuel Bauer
- a Department of Obstetrics and Gynecology , Beaumont Health , Royal Oak , MI , USA
| | - Zeynep Alpay-Savasan
- a Department of Obstetrics and Gynecology , Beaumont Health , Royal Oak , MI , USA
| | - Onur Turkoglu
- a Department of Obstetrics and Gynecology , Beaumont Health , Royal Oak , MI , USA
| | - Dotun Ogunyemi
- a Department of Obstetrics and Gynecology , Beaumont Health , Royal Oak , MI , USA
| | - Liona C Poon
- b Harris Birthright Research Centre for Fetal Medicine , Division of Women's Health, King's College Hospital , London , UK
| | - David S Wishart
- c Department of Biological Sciences , University of Alberta , Edmonton, Alberta , Canada , and.,d Department of Computing Sciences , University of Alberta , Edmonton, Alberta , Canada
| | - Kypros H Nicolaides
- b Harris Birthright Research Centre for Fetal Medicine , Division of Women's Health, King's College Hospital , London , UK
| |
Collapse
|
49
|
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]
|
50
|
|