1
|
Zhang L, Li W, Chi X, Sun Q, Li Y, Xing W, Ding G. Predictive performance of sFlt-1, PlGF and the sFlt-1/PlGF ratio for preeclampsia: A systematic review and meta-analysis. J Gynecol Obstet Hum Reprod 2025; 54:102925. [PMID: 39947348 DOI: 10.1016/j.jogoh.2025.102925] [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: 11/25/2023] [Revised: 01/15/2025] [Accepted: 02/08/2025] [Indexed: 02/19/2025]
Abstract
BACKGROUND It is difficult to evaluate whether monitoring serum sFlt-1, PlGF, or sFlt-1/PlGF in pregnant women who are suspected of having PE can significantly shorten the PE diagnosis time. OBJECTIVES To estimate the accuracy of sFlt-1, PlGF and sFlt-1/PlGF in preeclampsia prediction. SEARCH STRATEGY Databases including PubMed, Web of Science, Medline, CNKI, SinoMed, VIP Journal, and Wanfang Data were searched for eligible studies published until October 7, 2022. SELECTION CRITERIA The research subjects were pregnant women with or without PE. The research types were case-control studies and cohort studies. This was an original study involving the detection of at least one of the following in the blood, serum or plasma: sFlt-1, PlGF, and sFlt-1/PlGF. DATA COLLECTION AND ANALYSIS StataSE 16.0 was employed, using the Sen, Spe, PLR, and NLR to plot SROC, and subgroup analysis and meta-regression were conducted. MAIN RESULTS Meta-analysis showed that the combined Sen of sFlt, PlGF and sFlt-1/PlGF was 0.79 (95 % CI: 0.68-0.87), 0.76 (95 % CI: 0.69-0.82), and 0.83 (95 % CI: 0.77-0.88), respectively; the Spe was 0.86 (95 % CI: 0.77-0.92), 0.83 (95 % CI: 0.78-0.88), and 0.88 (95 % CI: 0.82-0.92), respectively; and the AUC was 0.89, 0.87, and 0.92, respectively. It was found to be attributable to study design, literature quality, sample size, disease subtypes, and cut-off values by using subgroup analysis and meta-regression. CONCLUSIONS The sFlt-1/PlGF ratio showed better predictive performance for preeclampsia than sFlt-1 or PlGF alone. However, the predictive value of the latter two cannot be ignored.This study highlights the performance of biomarkers in the diagnosis and prediction of PE, but that there is currently a lack of data to assess the value of using these biomarkers in clinical practice, and that the use of these biomarkers has not yet been shown to improve pregnancy outcomes.
Collapse
Affiliation(s)
- Luhan Zhang
- Xinjiang Clinical Research Center for Perinatal Diseases, Urumqi Maternal and Child Health Hospital, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Wenjing Li
- Xinjiang Clinical Research Center for Perinatal Diseases, Urumqi Maternal and Child Health Hospital, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Xiaolan Chi
- Xinjiang Clinical Research Center for Perinatal Diseases, Urumqi Maternal and Child Health Hospital, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Qi Sun
- Medical Research Design and Data Analysis Center, Traditional Chinese Medicine Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Yuanyuan Li
- Medical Research Design and Data Analysis Center, Traditional Chinese Medicine Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Weiwei Xing
- Medical Research Design and Data Analysis Center, Traditional Chinese Medicine Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Guifeng Ding
- Xinjiang Clinical Research Center for Perinatal Diseases, Urumqi Maternal and Child Health Hospital, Urumqi, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
2
|
Solt I, Cohen SM, Admati I, Beharier O, Dominsky O, Yagel S. Placenta at single-cell resolution in early and late preeclampsia: insights and clinical implications. Am J Obstet Gynecol 2025; 232:S176-S189. [PMID: 40253080 DOI: 10.1016/j.ajog.2025.01.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/31/2025] [Accepted: 01/31/2025] [Indexed: 04/21/2025]
Abstract
Preeclampsia, one of the great obstetrical syndromes, manifests through diverse maternal and fetal complications and remains a leading contributor to adverse perinatal outcomes. In this review, we describe our work on single-cell and single-nuclei RNA sequencing to elucidate the molecular mechanisms that underlie early- and late-onset preeclampsia. Analysis of 46 cell types, encompassing approximately 90,000 cells from placental tissues collected after delivery, demonstrated cellular dysregulation in early-onset preeclampsia, whereas late-onset preeclampsia showed comparatively subtle changes. These findings were observed in all cell lines, including all types of trophoblast, lymphoid, myeloid, stromal, and endothelial cells. Key findings in early-onset preeclampsia included disrupted syncytiotrophoblast and extravillous trophoblast angiogenic signaling, characterized by an up-regulation of FLT1 and down-regulation of PGF, consistent with an angiogenic imbalance. The stromal and vascular compartments exhibited stress-induced transcriptomic shifts. Both endothelial cells and pericytes showed evidence of stress, including up-regulation of heat shock proteins and markers of apoptosis. In addition, the inflammation- and stress-responsive states were more abundant in early-onset preeclampsia than in matched controls. Inflammatory pathways were markedly up-regulated in both the maternal and fetal immune cells; for example, we observed a marked increase in pro-inflammatory cytokines, including secreted phosphoprotein 1 and C-X-C motif chemokine ligand 2 and 3. Conversely, late-onset preeclampsia retained adaptive placental features with localized dysregulation of extracellular matrix remodeling and angiogenic markers, underscoring its possible maternal cardiovascular etiology. Single-cell and single-nuclei RNA sequencing investigations of placental tissues support the proposed classification of preeclampsia into a placental dysfunction type, primarily presenting early in pregnancy, and a maternal cardiovascular maladaptation type, primarily presenting later in pregnancy, each with distinct biomarkers, risk factors, and therapeutic targets. The early-onset preeclampsia findings advocate for interventions that target angiogenic pathways, such as RNA-based therapies that target specific cells of the placenta, to modulate soluble fms-like tyrosine kinase-1 levels. In contrast, late-onset preeclampsia management may benefit from maternal cardiovascular optimization, including individualized antihypertensive and metabolic treatments. These results underscore the heterogeneity of preeclampsia, emphasizing the need for individualized diagnostic and therapeutic strategies. This molecular atlas of preeclampsia advances our understanding of the complex interplay among elements of the maternal-placental-fetal array, thereby bridging clinical phenotypes and cellular mechanisms. Future research should focus on integrating these insights into longitudinal studies to develop precision medicine approaches for preeclampsia to enhance outcomes for mothers and neonates.
Collapse
Affiliation(s)
- Ido Solt
- Department of Obstetrics and Gynecology, Rambam Health Care Campus & Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Sarah M Cohen
- Division of Obstetrics and Gynecology, Hadassah Hebrew University Medical Centers, Jerusalem, Israel
| | - Inbal Admati
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology Haifa, Israel
| | - Ofer Beharier
- Division of Obstetrics and Gynecology, Hadassah Hebrew University Medical Centers, Jerusalem, Israel
| | - Omri Dominsky
- Department of Obstetrics and Gynecology, Lis Hospital for Women's Health Sourasky Medical Center, affiliated with the Faculty of Medicine at Tel Aviv University, Tel Aviv, Israel
| | - Simcha Yagel
- Division of Obstetrics and Gynecology, Hadassah Hebrew University Medical Centers, Jerusalem, Israel.
| |
Collapse
|
3
|
Lourenço J, Guedes-Martins L. Pathophysiology of Maternal Obesity and Hypertension in Pregnancy. J Cardiovasc Dev Dis 2025; 12:91. [PMID: 40137089 PMCID: PMC11942925 DOI: 10.3390/jcdd12030091] [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: 01/27/2025] [Revised: 02/22/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Obesity is one of the biggest health problems in the 21st century and the leading health disorder amongst women of fertile age. Maternal obesity is associated with several adverse maternal and fetal outcomes. In this group of women, the risk for the development of hypertensive disorders of pregnancy (HDPs), such as gestational hypertension (GH) and pre-eclampsia (PE), is increased. In fact, there is a linear association between an increase in pre-pregnancy body mass index (BMI) and PE. Excessive weight gain during pregnancy is also related to the development of PE and GH. The role of obesity in the pathophysiology of HDP is complex and is most likely due to an interaction between several factors that cause a state of poor maternal cardiometabolic health. Adipokines seem to have a central role in HDP development, especially for PE. Hypoadiponectinemia, hyperleptinemia, insulin resistance (IR), and a proinflammatory state are metabolic disturbances related to PE pathogenesis, contributing to its development by inducing a state of maternal endothelial dysfunction. Hypertriglyceridemia is suggested to also be a part of the disease mechanisms of HDP. Therefore, this review seeks to explore the scientific literature to assess the complications of maternal obesity and its association with the development of HDP.
Collapse
Affiliation(s)
- Joana Lourenço
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal;
- Centro de Medicina Fetal, Medicina Fetal Porto—Centro Materno Infantil do Norte, 4099-001 Porto, Portugal
| | - Luís Guedes-Martins
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal;
- Centro de Medicina Fetal, Medicina Fetal Porto—Centro Materno Infantil do Norte, 4099-001 Porto, Portugal
- Departamento da Mulher e da Medicina Reprodutiva, Serviço de Obstetrícia, Unidade Local de Saúde de Santo António EPE, Centro Materno Infantil do Norte, Largo Prof. Abel Salazar, 4099-001 Porto, Portugal
- Unidade de Investigação e Formação, Centro Materno Infantil do Norte, 4099-001 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-319 Porto, Portugal
| |
Collapse
|
4
|
Admati I, Skarbianskis N, Hochgerner H, Ophir O, Yagel S, Solt I, Zeisel A. Single-nuclei RNA-sequencing fails to detect molecular dysregulation in the preeclamptic placenta. Placenta 2025; 159:170-179. [PMID: 39733647 DOI: 10.1016/j.placenta.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/09/2024] [Accepted: 12/18/2024] [Indexed: 12/31/2024]
Abstract
INTRODUCTION Single-cell RNA-seq (scRNA-seq) revolutionized our understanding of tissue complexity in health and disease and revealed massive transcriptional dysregulation across placental cell classes in early-onset, but not late-onset preeclampsia (PE). However, the multinucleated syncytium is largely inaccessible to cell dissociation. Nuclei isolation and single-nuclei RNA-seq may be preferable in the placenta; not least considering compatibility with long-term tissue storage. Yet, nuclei contain a subsample of the cells' transcriptional profile. Mature transcripts critical to cellular function and disease may be missed. METHODS We analyzed placenta from pregnancies using single-cell and single-nuclei RNA-seq. The datasets comprise 45,836 cells and 27,078 nuclei, from 10 to 7 early-onset preeclampsia (EPE) cases and 3 and 2 early idiopathic controls (ECT), respectively. We compared the methods' sensitivities, cell type detection, differential gene expression in PE, and performed histological validations. RESULTS Mature syncytiotrophoblast were sampled ∼50x more efficiently after nuclei extraction. Yet, scRNA-seq was more sensitive in detection of genes, molecules and mature transcripts. In snRNA-seq, nuclei of all placental cell classes suffered ambient trophoblast contamination. Transcripts from extravillous trophoblast, stroma, vasculature and immune cells were profiled less comprehensively by single-nuclei RNA-seq (snRNA-seq), restricting cell-type detection. In EPE, we found dysregulation of angiogenic actors FLT1/PGF both in prefused syncytiotrophoblast after cell extraction, and mature syncytiotrophoblast after nuclei isolation. Disease-related stress and inflammation were undetected from nuclei. DISCUSSION scRNA-seq has important advantages over snRNA-seq for comprehensive transcriptomics studies of the placenta, especially to understand cell-type resolved dysregulation in pathologies. Yet, to address the dilemma of an underrepresented syncytium, studies benefit from complementary nuclei extraction.
Collapse
Affiliation(s)
- Inbal Admati
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Niv Skarbianskis
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Hannah Hochgerner
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Osnat Ophir
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Simcha Yagel
- Division of Obstetrics and Gynecology Hadassah, Hebrew University Medical Centers, Jerusalem, Israel
| | - Ido Solt
- Department of Obstetrics and Gynecology, Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel.
| | - Amit Zeisel
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
5
|
Andresen IJ, Zucknick M, Degnes MHL, Angst MS, Aghaeepour N, Romero R, Roland MCP, Tarca AL, Westerberg AC, Michelsen TM. Prediction of late-onset preeclampsia using plasma proteomics: a longitudinal multi-cohort study. Sci Rep 2024; 14:30813. [PMID: 39730472 DOI: 10.1038/s41598-024-81277-2] [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: 08/27/2024] [Accepted: 11/22/2024] [Indexed: 12/29/2024] Open
Abstract
Preeclampsia is a pregnancy disorder with substantial perinatal and maternal morbidity and mortality. Pregnant women at risk of preeclampsia would benefit from early detection for follow-up, timely interventions and delivery. Several attempts have been made to identify protein biomarkers of preeclampsia, but findings vary with demographics, clinical characteristics, and time of sampling. In the current study, we combined three independent longitudinal pregnancy cohorts (Detroit, Stanford and Oslo) resulting in 124 late-onset preeclampsia (LOPE) cases and 178 gestational age matched controls, and analyzed > 1000 proteins in maternal plasma sampled between 12 and 34 weeks of gestation. Differential abundance analysis of combined protein data revealed increased deviation in protein abundance trajectories throughout gestation in women destined to develop LOPE compared to controls. There were no differentially abundant proteins at time interval T1 (12-19 weeks), yet 31 differentially abundant proteins were found at time interval T2 (19-27 weeks), and 48 proteins at time interval T3 (27- 34 weeks). Multi-protein random forest models assessed via cross-validation predicted LOPE with an area under the ROC curve of 0.72 (0.65-0.78), 0.76 (0.71-0.81) and 0.80 (0.75-0.85) at time interval T1, T2 and T3, respectively. The results at T3 were confirmed using a leave-one-cohort-out analysis suggesting cross-cohort consistency, and at T1 and T2 when the largest two cohorts were used as training sets.
Collapse
Affiliation(s)
- Ina J Andresen
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital Rikshospitalet, Oslo, Norway.
| | - Manuela Zucknick
- Oslo Centre for Biostatistics and Epidemiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Maren-Helene L Degnes
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, US Department of Health and Human Services (NICHD/NIH/DHHS), Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, MD and Detroit, Bethesda, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Marie Cecilie P Roland
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Adi L Tarca
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Ane Cecilie Westerberg
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital Rikshospitalet, Oslo, Norway
- School of Health Sciences , Kristiania University College , Oslo, Norway
| | - Trond M Michelsen
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital Rikshospitalet, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
6
|
Suksai M, Romero R, Bosco M, Gotsch F, Jung E, Chaemsaithong P, Tarca AL, Gudicha DW, Gomez-Lopez N, Arenas-Hernandez M, Meyyazhagan A, Grossman LI, Aras S, Chaiworapongsa T. A mitochondrial regulator protein, MNRR1, is elevated in the maternal blood of women with preeclampsia. J Matern Fetal Neonatal Med 2024; 37:2297158. [PMID: 38220225 DOI: 10.1080/14767058.2023.2297158] [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: 06/29/2023] [Accepted: 12/15/2023] [Indexed: 01/16/2024]
Abstract
OBJECTIVE Preeclampsia, one of the most serious obstetric complications, is a heterogenous disorder resulting from different pathologic processes. However, placental oxidative stress and an anti-angiogenic state play a crucial role. Mitochondria are a major source of cellular reactive oxygen species. Abnormalities in mitochondrial structures, proteins, and functions have been observed in the placentae of patients with preeclampsia, thus mitochondrial dysfunction has been implicated in the mechanism of the disease. Mitochondrial nuclear retrograde regulator 1 (MNRR1) is a newly characterized bi-organellar protein with pleiotropic functions. In the mitochondria, this protein regulates cytochrome c oxidase activity and reactive oxygen species production, whereas in the nucleus, it regulates the transcription of a number of genes including response to tissue hypoxia and inflammatory signals. Since MNRR1 expression changes in response to hypoxia and to an inflammatory signal, MNRR1 could be a part of mitochondrial dysfunction and involved in the pathologic process of preeclampsia. This study aimed to determine whether the plasma MNRR1 concentration of women with preeclampsia differed from that of normal pregnant women. METHODS This retrospective case-control study included 97 women with preeclampsia, stratified by gestational age at delivery into early (<34 weeks, n = 40) and late (≥34 weeks, n = 57) preeclampsia and by the presence or absence of placental lesions consistent with maternal vascular malperfusion (MVM), the histologic counterpart of an anti-angiogenic state. Women with an uncomplicated pregnancy at various gestational ages who delivered at term served as controls (n = 80) and were further stratified into early (n = 25) and late (n = 55) controls according to gestational age at venipuncture. Maternal plasma MNRR1 concentrations were determined by an enzyme-linked immunosorbent assay. RESULTS 1) Women with preeclampsia at the time of diagnosis (either early or late disease) had a significantly higher median (interquartile range, IQR) plasma MNRR1 concentration than the controls [early preeclampsia: 1632 (924-2926) pg/mL vs. 630 (448-4002) pg/mL, p = .026, and late preeclampsia: 1833 (1441-5534) pg/mL vs. 910 (526-6178) pg/mL, p = .021]. Among women with early preeclampsia, those with MVM lesions in the placenta had the highest median (IQR) plasma MNRR1 concentration among the three groups [with MVM: 2066 (1070-3188) pg/mL vs. without MVM: 888 (812-1781) pg/mL, p = .03; and with MVM vs. control: 630 (448-4002) pg/mL, p = .04]. There was no significant difference in the median plasma MNRR1 concentration between women with early preeclampsia without MVM lesions and those with an uncomplicated pregnancy (p = .3). By contrast, women with late preeclampsia, regardless of MVM lesions, had a significantly higher median (IQR) plasma MNRR1 concentration than women in the control group [with MVM: 1609 (1392-3135) pg/mL vs. control: 910 (526-6178), p = .045; and without MVM: 2023 (1578-8936) pg/mL vs. control, p = .01]. CONCLUSIONS MNRR1, a mitochondrial regulator protein, is elevated in the maternal plasma of women with preeclampsia (both early and late) at the time of diagnosis. These findings may reflect some degree of mitochondrial dysfunction, intravascular inflammation, or other unknown pathologic processes that characterize this obstetrical syndrome.
Collapse
Affiliation(s)
- Manaphat Suksai
- Pregnancy 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, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Roberto Romero
- Pregnancy 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, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Mariachiara Bosco
- Pregnancy 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, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Obstetrics and Gynecology, AOUI Verona, University of Verona, Verona, Italy
| | - Francesca Gotsch
- Pregnancy 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, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Eunjung Jung
- Pregnancy 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, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Piya Chaemsaithong
- Pregnancy 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, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Adi L Tarca
- Pregnancy 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, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Dereje W Gudicha
- Pregnancy 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, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nardhy Gomez-Lopez
- Pregnancy 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, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Marcia Arenas-Hernandez
- Pregnancy 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, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Arun Meyyazhagan
- Pregnancy 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, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Centre of Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| | - Lawrence I Grossman
- Pregnancy 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, United States Department of Health and Human Services, Bethesda, MD, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Siddhesh Aras
- Pregnancy 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, United States Department of Health and Human Services, Bethesda, MD, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Tinnakorn Chaiworapongsa
- Pregnancy 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, United States Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
7
|
Westerberg AC, Degnes MHL, Andresen IJ, Roland MCP, Michelsen TM. Angiogenic and vasoactive proteins in the maternal-fetal interface in healthy pregnancies and preeclampsia. Am J Obstet Gynecol 2024; 231:550.e1-550.e22. [PMID: 38494070 DOI: 10.1016/j.ajog.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/08/2024] [Accepted: 03/08/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Preeclampsia is characterized by maternal endothelial activation and placental dysfunction. Imbalance in maternal angiogenic and vasoactive factors has been linked to the pathophysiology. The contribution of the placenta as a source of these factors remains unclear. Furthermore, little is known about fetal angiogenic and vasoactive proteins and the relation between maternal and fetal levels. OBJECTIVE We describe placental growth factor, soluble Fms-like tyrosine kinase 1, soluble endoglin, and endothelin 1-3 in 5 vessels in healthy pregnancies, early- and late-onset preeclampsia. Specifically, we aimed to (1) compare protein abundance in vessels at the maternal-fetal interface between early- and late-onset preeclampsia, and healthy pregnancies, (2) describe placental uptake and release of proteins, and (3) describe protein abundance in the maternal vs fetal circulations. STUDY DESIGN Samples were collected from the maternal radial artery, uterine vein and antecubital vein, and fetal umbilical vein and artery in 75 healthy and 37 preeclamptic mother-fetus pairs (including 19 early-onset preeclampsia and 18 late-onset preeclampsia), during scheduled cesarean delivery. This method allows estimation of placental release and uptake of proteins by calculation of venoarterial differences on each side of the placenta. The microarray-based SomaScan assay quantified the proteins. RESULTS The abundance of soluble Fms-like tyrosine kinase 1 and endothelin 1 was higher in the maternal vessels in preeclampsia than in healthy pregnancies, with the highest abundance in early-onset preeclampsia. Placental growth factor was lower in the maternal vessels in early-onset preeclampsia than in both healthy and late-onset preeclampsia. Maternal endothelin 2 was higher in preeclampsia, with late-onset preeclampsia having the highest abundance. Our model confirmed placental release of placental growth factor and soluble Fms-like tyrosine kinase 1 to the maternal circulation in all groups. The placenta released soluble Fms-like tyrosine kinase 1 into the fetal circulation in healthy and late-onset preeclampsia pregnancies. Fetal endothelin 1 and soluble Fms-like tyrosine kinase 1 were higher in early-onset preeclampsia, whereas soluble endoglin and endothelin 3 were lower in both preeclampsia groups than healthy controls. Across groups, abundances of placental growth factor, soluble Fms-like tyrosine kinase 1, and endothelin 3 were higher in the maternal artery than the fetal umbilical vein, whereas endothelin 2 was lower. CONCLUSION An increasing abundance of maternal soluble Fms-like tyrosine kinase 1 and endothelin 1 across the groups healthy, late-onset preeclampsia and early-onset combined with a positive correlation may suggest that these proteins are associated with the pathophysiology and severity of the disease. Elevated endothelin 1 in the fetal circulation in early-onset preeclampsia represents a novel finding. The long-term effects of altered protein abundance in preeclampsia on fetal development and health remain unknown. Further investigation of these proteins' involvement in the pathophysiology and as treatment targets is warranted.
Collapse
Affiliation(s)
- Ane Cecilie Westerberg
- Division of Obstetrics and Gynecology, Department of Obstetrics, Oslo University Hospital Rikshospitalet, Oslo, Norway; School of Health Sciences, Kristiania University College, Oslo, Norway.
| | - Maren-Helene Langeland Degnes
- Division of Obstetrics and Gynecology, Department of Obstetrics, Oslo University Hospital Rikshospitalet, Oslo, Norway; Department of Biostatistics, Oslo Centre for Biostatistics and Epidemiology, University of Oslo, Oslo, Norway
| | - Ina Jungersen Andresen
- Division of Obstetrics and Gynecology, Department of Obstetrics, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Marie Cecilie Paasche Roland
- Division of Obstetrics and Gynecology, Department of Obstetrics, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Trond Melbye Michelsen
- Division of Obstetrics and Gynecology, Department of Obstetrics, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
8
|
Khalil A, Bellesia G, Norton ME, Jacobsson B, Haeri S, Egbert M, Malone FD, Wapner RJ, Roman A, Faro R, Madankumar R, Strong N, Silver RM, Vohra N, Hyett J, MacPherson C, Prigmore B, Ahmed E, Demko Z, Ortiz JB, Souter V, Dar P. The role of cell-free DNA biomarkers and patient data in the early prediction of preeclampsia: an artificial intelligence model. Am J Obstet Gynecol 2024; 231:554.e1-554.e18. [PMID: 38432413 DOI: 10.1016/j.ajog.2024.02.299] [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: 05/12/2023] [Revised: 02/16/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Accurate individualized assessment of preeclampsia risk enables the identification of patients most likely to benefit from initiation of low-dose aspirin at 12 to 16 weeks of gestation when there is evidence for its effectiveness, and enables the guidance of appropriate pregnancy care pathways and surveillance. OBJECTIVE The primary objective of this study was to evaluate the performance of artificial neural network models for the prediction of preterm preeclampsia (<37 weeks' gestation) using patient characteristics available at the first antenatal visit and data from prenatal cell-free DNA screening. Secondary outcomes were prediction of early-onset preeclampsia (<34 weeks' gestation) and term preeclampsia (≥37 weeks' gestation). METHODS This secondary analysis of a prospective, multicenter, observational prenatal cell-free DNA screening study (SMART) included singleton pregnancies with known pregnancy outcomes. Thirteen patient characteristics that are routinely collected at the first prenatal visit and 2 characteristics of cell-free DNA (total cell-free DNA and fetal fraction) were used to develop predictive models for early-onset (<34 weeks), preterm (<37 weeks), and term (≥37 weeks) preeclampsia. For the models, the "reference" classifier was a shallow logistic regression model. We also explored several feedforward (nonlinear) neural network architectures with ≥1 hidden layers, and compared their performance with the logistic regression model. We selected a simple neural network model built with 1 hidden layer and made up of 15 units. RESULTS Of the 17,520 participants included in the final analysis, 72 (0.4%) developed early-onset, 251 (1.4%) preterm, and 420 (2.4%) term preeclampsia. Median gestational age at cell-free DNA measurement was 12.6 weeks, and 2155 (12.3%) had their cell-free DNA measurement at ≥16 weeks' gestation. Preeclampsia was associated with higher total cell-free DNA (median, 362.3 vs 339.0 copies/mL cell-free DNA; P<.001) and lower fetal fraction (median, 7.5% vs 9.4%; P<.001). The expected, cross-validated area under the curve scores for early-onset, preterm, and term preeclampsia were 0.782, 0.801, and 0.712, respectively, for the logistic regression model, and 0.797, 0.800, and 0.713, respectively, for the neural network model. At a screen-positive rate of 15%, sensitivity for preterm preeclampsia was 58.4% (95% confidence interval, 0.569-0.599) for the logistic regression model and 59.3% (95% confidence interval, 0.578-0.608) for the neural network model. The contribution of both total cell-free DNA and fetal fraction to the prediction of term and preterm preeclampsia was negligible. For early-onset preeclampsia, removal of the total cell-free DNA and fetal fraction features from the neural network model was associated with a 6.9% decrease in sensitivity at a 15% screen-positive rate, from 54.9% (95% confidence interval, 52.9-56.9) to 48.0% (95% confidence interval, 45.0-51.0). CONCLUSION Routinely available patient characteristics and cell-free DNA markers can be used to predict preeclampsia with performance comparable to that of other patient characteristic models for the prediction of preterm preeclampsia. Logistic regression and neural network models showed similar performance.
Collapse
Affiliation(s)
- Asma Khalil
- Department of Obstetrics and Gynaecology, St. George's Hospital, St. George's University of London, London, United Kingdom.
| | | | - Mary E Norton
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California San Francisco, San Francisco, CA
| | - Bo Jacobsson
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Sina Haeri
- Austin Maternal-Fetal Medicine, Austin, TX
| | | | - Fergal D Malone
- Department of Obstetrics and Gynaecology, Rotunda Hospital, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ronald J Wapner
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY
| | - Ashley Roman
- Department of Obstetrics and Gynecology, New York University Grossman School of Medicine, New York, NY
| | - Revital Faro
- Department of Obstetrics and Gynecology, Saint Peter's University Hospital, New Brunswick, NJ
| | - Rajeevi Madankumar
- Department of Obstetrics and Gynecology, Long Island Jewish Medical Center, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, NY
| | - Noel Strong
- Department of Obstetrics and Gynecology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Robert M Silver
- Department of Obstetrics and Gynecology, University of Utah, Salt Lake City, UT
| | - Nidhi Vohra
- Department of Obstetrics and Gynecology, North Shore University Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
| | - Jon Hyett
- Department of Obstetrics and Gynaecology, Royal Prince Alfred Hospital, Western Sydney University, Sydney, Australia
| | - Cora MacPherson
- Biostatistics Center, George Washington University, Rockville, MD
| | | | | | | | | | | | - Pe'er Dar
- Department of Obstetrics and Gynecology and Women's Health, Montefiore Medical Center, Albert Einstein College of Medicine, New York, NY
| |
Collapse
|
9
|
Bucher V, Mitchell AR, Gudmundsson P, Atkinson J, Wallin N, Asp J, Sennström M, Hildén K, Edvinsson C, Ek J, Hastie R, Cluver C, Bergman L. Prediction of adverse maternal and perinatal outcomes associated with pre-eclampsia and hypertensive disorders of pregnancy: a systematic review and meta-analysis. EClinicalMedicine 2024; 76:102861. [PMID: 39391014 PMCID: PMC11465897 DOI: 10.1016/j.eclinm.2024.102861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024] Open
Abstract
Background Hypertensive disorders of pregnancy are a leading cause of maternal and perinatal morbidity and mortality. If women at high risk for developing complications could be identified early, level of care could be triaged, limited resources could be correctly allocated and targeted interventions to prevent complications could be implemented. Methods We updated a systematic review and meta-analysis and added single outcomes. Women with hypertensive disorders of pregnancy were included. Exposures were tests predicting adverse maternal and/or perinatal outcomes. We searched Medline, Embase, CINAHL, and Cochrane library from January 2016-February 2024. We included studies identified from the previous review. We calculated effect measures. For similar predictive tests and outcomes, area under the receiver-operating-characteristic curve (AUROC) were pooled. This study was registered by PROSPERO: CRD42022336368. Findings Of the 2898 studies identified, 80 were included. Thirty were added from the previous review resulting in 110 included studies with 506,178 women. Despite more than 1500 tests being performed, most outcomes could not be pooled due to heterogeneity in populations, tests, and outcome definitions. For maternal outcomes, only studies reporting on the Pre-eclampsia Integrated Estimate of RiSk (fullPIERS) model could be pooled. For the composite outcome within 48-h the AUROC was 0.78 (95% CI 0.71-0.86, N = 8). There was significant heterogeneity (I 2 = 95.7%). For perinatal outcomes, data were pooled for pulsatility index in the umbilical artery and soluble FMS-like tyrosine kinase-1 (sFlt-1)/placental growth factor (PlGF) ratio. Biomarkers like the sFlt-1/PlGF ratio showed promising predictive performance for some outcomes but were not externally validated. Interpretation Despite including over 100 studies with more than 1500 predictors, we were unable to pool any single maternal outcomes and only a few individual perinatal outcomes. The fullPIERS model was externally validated, showing moderate accuracy which varied across studies and should be validated in each new population. Angiogenic biomarkers showed promise but need validation. Future studies should use standardized outcome measures and validate promising tests. Funding VB is supported by the Swedish Research Council, Grant number 2020-01481. University of Gothenburg.
Collapse
Affiliation(s)
- Valentina Bucher
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alexandra Roddy Mitchell
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Melbourne, Australia
| | - Pia Gudmundsson
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Obstetrics and Gynecology, Gothenburg, Sweden
| | - Jessica Atkinson
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Melbourne, Australia
| | - Nicole Wallin
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Obstetrics and Gynecology, Gothenburg, Sweden
| | - Joline Asp
- Department of Women’s and Children’s Health, Clinical Obstetrics, Uppsala University, Uppsala, Sweden
| | - Maria Sennström
- Department of Women's and Children's Health, Division for Obstetrics and Gynecology, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Karin Hildén
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Camilla Edvinsson
- Department of Obstetrics and Gynaecology, Institute of Clinical Sciences, Lund University, Lund, Sweden
| | - Joakim Ek
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Roxanne Hastie
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Melbourne, Australia
- Department of Women’s and Children’s Health, Clinical Obstetrics, Uppsala University, Uppsala, Sweden
- Department of Obstetrics and Gynecology, Stellenbosch University, Cape Town, South Africa
| | - Catherine Cluver
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Melbourne, Australia
- Department of Obstetrics and Gynecology, Stellenbosch University, Cape Town, South Africa
| | - Lina Bergman
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Obstetrics and Gynecology, Gothenburg, Sweden
- Department of Women’s and Children’s Health, Clinical Obstetrics, Uppsala University, Uppsala, Sweden
- Department of Obstetrics and Gynecology, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
10
|
Than NG, Romero R, Fitzgerald W, Gudicha DW, Gomez-Lopez N, Posta M, Zhou F, Bhatti G, Meyyazhagan A, Awonuga AO, Chaiworapongsa T, Matthies D, Bryant DR, Erez O, Margolis L, Tarca AL. Proteomic Profiles of Maternal Plasma Extracellular Vesicles for Prediction of Preeclampsia. Am J Reprod Immunol 2024; 92:e13928. [PMID: 39347565 PMCID: PMC12087260 DOI: 10.1111/aji.13928] [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] [Received: 02/15/2024] [Revised: 08/30/2024] [Accepted: 09/01/2024] [Indexed: 10/01/2024] Open
Abstract
PROBLEM Preeclampsia is a heterogeneous syndrome of diverse etiologies and molecular pathways leading to distinct clinical subtypes. Herein, we aimed to characterize the extracellular vesicle (EV)-associated and soluble fractions of the maternal plasma proteome in patients with preeclampsia and to assess their value for disease prediction. METHOD OF STUDY This case-control study included 24 women with term preeclampsia, 23 women with preterm preeclampsia, and 94 healthy pregnant controls. Blood samples were collected from cases on average 7 weeks before the diagnosis of preeclampsia and were matched to control samples. Soluble and EV fractions were separated from maternal plasma; EVs were confirmed by cryo-EM, NanoSight, and flow cytometry; and 82 proteins were analyzed with bead-based, multiplexed immunoassays. Quantile regression analysis and random forest models were implemented to evaluate protein concentration differences and their predictive accuracy. Preeclampsia subgroups defined by molecular profiles were identified by hierarchical cluster analysis. Significance was set at p < 0.05 or false discovery rate-adjusted q < 0.1. RESULTS In preterm preeclampsia, PlGF, PTX3, and VEGFR-1 displayed differential abundance in both soluble and EV fractions, whereas angiogenin, CD40L, endoglin, galectin-1, IL-27, CCL19, and TIMP1 were changed only in the soluble fraction (q < 0.1). The direction of changes in the EV fraction was consistent with that in the soluble fraction for nine proteins. In term preeclampsia, CCL3 had increased abundance in both fractions (q < 0.1). The combined EV and soluble fraction proteomic profiles predicted preterm and term preeclampsia with an AUC of 78% (95% CI, 66%-90%) and 68% (95% CI, 56%-80%), respectively. Three clusters of preeclampsia featuring distinct clinical characteristics and placental pathology were identified based on combined protein data. CONCLUSIONS Our findings reveal distinct alterations of the maternal EV-associated and soluble plasma proteome in preterm and term preeclampsia and identify molecular subgroups of patients with distinct clinical and placental histopathologic features.
Collapse
Affiliation(s)
- Nándor Gábor Than
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
- Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary
| | - Roberto Romero
- Pregnancy 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, 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
| | - Wendy Fitzgerald
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Dereje W. Gudicha
- Pregnancy 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, USA
| | - Nardhy Gomez-Lopez
- Department of Obstetrics and Gynecology & Department of Pathology and Immunology, Washington University, St. Louis, Missouri, USA
| | - Máté Posta
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University Doctoral School, Budapest, Hungary
| | - Fei Zhou
- Unit on Structural Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Gaurav Bhatti
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Arun Meyyazhagan
- Pregnancy 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, USA
| | - Awoniyi O. Awonuga
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Tinnakorn Chaiworapongsa
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Doreen Matthies
- Unit on Structural Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - David R. Bryant
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Offer Erez
- Department of Obstetrics and Gynecology, Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Leonid Margolis
- Faculty of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia
| | - Adi L. Tarca
- Center for Molecular Medicine and Genetics, Wayne State University, 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
| |
Collapse
|
11
|
Arbuzova S. Common pathogenesis of early and late preeclampsia: evidence from recurrences and review of the literature. Arch Gynecol Obstet 2024; 310:953-959. [PMID: 37740793 PMCID: PMC11258074 DOI: 10.1007/s00404-023-07217-z] [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: 07/27/2023] [Accepted: 09/03/2023] [Indexed: 09/25/2023]
Abstract
OBJECTIVE To investigate whether there is an association between the gestational age at the onset of preeclampsia in recurrent cases and the gestational age at the onset of preeclampsia in previous pregnancies. METHODS This retrospective nested case-control study was designed to investigate whether gestational age at diagnosis and at delivery in recurrent cases of preeclampsia correlates with gestational age at diagnosis and delivery in the previous cases of preeclampsia in the same individuals. The database of a Ukrainian research network was used to find patients with the diagnosis of preeclampsia between 2019 and 2021. The database was further queried to identify those with a history of preeclampsia in a previous pregnancy. The comparison was made using the Pearson correlation coefficient. RESULTS One hundred and three patients who were diagnosed with preeclampsia were identified. Of those, 15 had recurrent preeclampsia, 2 of whom had preeclampsia in 2 previous pregnancies. There was no statistically significant correlation: based on gestational age at delivery R = - 0.28 (P = 0.30; 95% confidence interval (- 0.69 to 0.28) and based on gestational age at the time of diagnosis R = - 0.14 (P = 0.62; - 0.60 to 0.41). CONCLUSION Our data do not find an association between the gestational age of recurrent preeclampsia and preeclampsia diagnosed in a previous pregnancy. This supports the idea that there is single pathogenesis for preeclampsia regardless of the gestational age. It suggests that there are variations in the course of preeclampsia that may be determined by the capacity of the compensatory mechanisms.
Collapse
Affiliation(s)
- Svitlana Arbuzova
- Eastern-Ukrainian Center for Medical Genetics and Prenatal Diagnosis, Mariupol, Kiev, Ukraine.
- Institute of Health Research, University of Exeter, Studio 3.4, Block M, Birks Hall, New North Road, Exeter, Devon, EX4 4GH, England, UK.
| |
Collapse
|
12
|
Hunt BL, Parikh A, Jain D. The Factors Associated With Continuous Positive Airway Pressure (CPAP) Failure in Late Preterm and Term Infants and Its Impact on In-Hospital Outcomes. Cureus 2024; 16:e63895. [PMID: 39100029 PMCID: PMC11298116 DOI: 10.7759/cureus.63895] [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] [Accepted: 07/05/2024] [Indexed: 08/06/2024] Open
Abstract
Background and objective Late preterm and term infants commonly require continuous positive airway pressure (CPAP) on admission. However, CPAP failure in this population has not been well studied. Hence, we conducted this study to determine the impact of CPAP failure and identify antenatal factors associated with it in late preterm and term infants. Materials and methods We carried out a single-center retrospective analysis of all inborn infants of ≥34 weeks gestational age (GA) from 2012 to 2019 who received CPAP on admission to the neonatal intensive care unit (NICU). CPAP failure was defined as follows: escalation in the mode of respiratory support, surfactant administration, increase in FiO2 >0.2 above the baseline, or absolute FiO2 >0.4 for ≥3h; within 12h of admission. In-hospital outcomes and perinatal factors were compared between CPAP-failure and success groups. Multivariate stepwise binary logistic regression analysis (LRA) was used to assess the association between antenatal factors and CPAP failure. Results Of the 272 infants included in the study, 38 (14%) failed CPAP. Infants in the failure group received a longer duration of respiratory support [median (IQR): 3.0 (5.6) vs. 0.5 (0.5)d; p<0.001], and length of stay [9 (9) vs. 4 (4)d; p<0.001]. On LRA, higher GA was associated with reduced odds of CPAP failure. Maternal hypertensive disorders, meconium-stained amniotic fluid, and group B Streptococcus (GBS)-positive status were associated with increased odds of CPAP failure. Conclusions In this cohort of late preterm and term infants, CPAP failure was associated with worse in-hospital outcomes. Lower GA, maternal hypertensive disorders, meconium-stained amniotic fluid, and GBS-positive status were associated with CPAP failure. These data, if replicated in further studies, may help develop individualized respiratory support strategies.
Collapse
Affiliation(s)
- Bethany L Hunt
- Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, USA
| | - Amy Parikh
- Pediatrics, Nationwide Children's Hospital, Columbus, USA
| | - Deepak Jain
- Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, USA
| |
Collapse
|
13
|
Roberts JM. Preeclampsia epidemiology(ies) and pathophysiology(ies). Best Pract Res Clin Obstet Gynaecol 2024; 94:102480. [PMID: 38490067 DOI: 10.1016/j.bpobgyn.2024.102480] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/14/2023] [Accepted: 02/05/2024] [Indexed: 03/17/2024]
Abstract
Preeclampsia/eclampsia was first described 2000 years ago. Concepts guiding diagnosis have changed over time making longitudinal studies challenging. Similarly, concepts of pathophysiology have evolved from eclampsia as a pregnancy seizure disorder to preeclampsia as a hypertensive and renal disorder to our current concept of a preeclampsia as a pregnancy specific, multisystemic inflammatory disorder. Although preeclampsia is pregnancy specific and many pathophysiologic findings begin to resolve with delivery, its impact extends beyond pregnancy. The risk of cardiovascular and neurological disease is increased after pregnancy in women who have had preeclampsia. The disorder is not a disease, but a syndrome and emerging data indicate multiple pathways to the syndrome. It is likely that our failure to have a major impact on prediction and prevention despite a large increase in understanding is due to the existence of multiple subtypes of preeclampsia. This concept should guide future research.
Collapse
Affiliation(s)
- James M Roberts
- Obstetrics Gynecology and Reproductive Sciences, Epidemiology and Clinical and Translational Research University of Pittsburgh, 10 Georgian Place, Pittsburgh, PA, 15215, United States.
| |
Collapse
|
14
|
CHAIWORAPONGSA T, ROMERO R, GOMEZ-LOPEZ N, SUKSAI M, GALLO DM, JUNG E, BERRY SM, AWONUGA A, TARCA AL, BRYANT DR. Preeclampsia at term: evidence of disease heterogeneity based on the profile of circulating cytokines and angiogenic factors. Am J Obstet Gynecol 2024; 230:450.e1-450.e18. [PMID: 37806612 PMCID: PMC10990810 DOI: 10.1016/j.ajog.2023.10.002] [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: 08/08/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Intravascular inflammation and an antiangiogenic state have been implicated in the pathophysiology of preeclampsia. On the basis of the profiles of their angiogenic/antiangiogenic factors, women with preeclampsia at term may be classified into 2 subgroups with different characteristics and prevalence of adverse outcomes. This study was undertaken to examine whether these 2 subgroups of preeclampsia at term also show differences in their profiles of intravascular inflammation. OBJECTIVE This study aimed to determine the plasma profiles of cytokines and chemokines in women with preeclampsia at term who had a normal or an abnormal angiogenic profile. STUDY DESIGN A nested case-control study was conducted to include women classified into 3 groups: women with an uncomplicated pregnancy (n=213) and women with preeclampsia at term with a normal (n=55) or an abnormal (n=41) angiogenic profile. An abnormal angiogenic profile was defined as a plasma ratio of placental growth factor and soluble fms-like tyrosine kinase-1 multiple of the median <10th percentile for gestational age. Concentrations of cytokines were measured by multiplex immunoassays. RESULTS Women with preeclampsia at term and an abnormal angiogenic profile showed evidence of the greatest intravascular inflammation among the study groups. These women had higher plasma concentrations of 5 cytokines (interleukin-6, interleukin-8, interleukin-12/interleukin-23p40, interleukin-15, and interleukin-16) and 7 chemokines (eotaxin, eotaxin-3, interferon-γ inducible protein-10, monocyte chemotactic protein-4, macrophage inflammatory protein-1β, macrophage-derived chemokine, and thymus and activation-regulated chemokine compared to women with an uncomplicated pregnancy. By contrast, women with preeclampsia at term and a normal angiogenic profile, compared to women with an uncomplicated pregnancy, had only a higher plasma concentration of monocyte chemotactic protein-4. A correlation between severity of the antiangiogenic state, blood pressure, and plasma concentrations of a subset of cytokines was observed. CONCLUSION Term preeclampsia can be classified into 2 clusters. One is characterized by an antiangiogenic state coupled with an excessive inflammatory process, whereas the other has neither of these features. These findings further support the heterogeneity of preeclampsia at term and may explain the distinct clinical outcomes.
Collapse
Affiliation(s)
- Tinnakorn CHAIWORAPONGSA
- Pregnancy 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, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto ROMERO
- Pregnancy 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, Bethesda, Maryland, 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
| | - Nardhy GOMEZ-LOPEZ
- Pregnancy 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, Bethesda, Maryland, 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
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Manaphat SUKSAI
- Pregnancy 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, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Dahiana M. GALLO
- Pregnancy 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, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Valle, Cali, Colombia
| | - Eunjung JUNG
- Pregnancy 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, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Stanley M. BERRY
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Awoniyi AWONUGA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Adi L. TARCA
- Pregnancy 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, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, USA
| | - David R. BRYANT
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
15
|
de Ganzo Suárez T, de Paco Matallana C, Plasencia W. Spiral, uterine artery doppler and placental ultrasound in relation to preeclampsia. Best Pract Res Clin Obstet Gynaecol 2024; 92:102426. [PMID: 38039843 DOI: 10.1016/j.bpobgyn.2023.102426] [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: 05/02/2023] [Revised: 06/18/2023] [Accepted: 08/06/2023] [Indexed: 12/03/2023]
Abstract
Preeclampsia (PE) is a multiorgan disorder that complicates around 2-8% of pregnancies and is a major cause of perinatal and maternal morbidity and mortality. PE is a clinical syndrome characterized by hypertension secondary to systemic inflammation, endothelial dysfunction, and syncytiotrophoblast stress leading to hypertension and multiorgan dysfunction. The uterine arteries are the main blood vessels that supply blood to the uterus. They give off branches and plays an important role in maintaining blood supply during pregnancy. The arcuate artery originates from the uterine artery and runs medially through the myometrium. The arcuate arteries divide almost directly into anterior and posterior branches, from which the radial artery leads directly to the uterine cavity during their course. Near the endometrium-myometrium junction, the radial artery generates spiral arteries within the basal layer and functional endometrium. The walls of radial and spiral arteries are rich in smooth muscle, which is lost when trophoblast cells invade and become large-caliber vessels. This physiological transformation of uteroplacental spiral arteries is critical for successful placental implantation and normal placental function. In normal pregnancy, the luminal diameter of the spiral arteries is greatly increased, and the vascular smooth muscle is replaced by trophoblast cells. This process and changes in the spiral arteries are called spiral artery remodeling. In PE, this genetically and immunologically governed process is deficient and therefore there is decreased vascular capacitance and increased resistance in the uteroplacental circulation. Furthermore, this defect in uteroplacental spiral artery remodeling is not only associated with early onset PE, but also with fetal growth restriction, placental abruption, and spontaneous premature rupture of membranes. Doppler ultrasound allows non-invasive assessment of placentation, while the flow impedance decreases as the pregnancy progresses in normal pregnancies, in those destined to develop preeclampsia the impedance is increased.
Collapse
Affiliation(s)
- Tania de Ganzo Suárez
- Department of Obstetrics and Gynecology, Complejo Hospitalario Universitario Nuestra Señora de la Candelaria, Tenerife, Canary Islands, Spain.
| | - Catalina de Paco Matallana
- Department of Obstetrics and Gynecology, Hospital Clínico Universitario Virgen de la Arrixaca, El Palmar, Murcia, Spain; Institute for Biomedical Research of Murcia, IMIB-Arrixaca, El Palmar, Murcia, Spain; Faculty of Medicine, Universidad de Murcia, Murcia, Spain.
| | - Walter Plasencia
- Department of Obstetrics and Gynecology, Complejo Hospitalario Universitario de Canarias, Tenerife. Canary Islands, Spain.
| |
Collapse
|
16
|
Boulanger H, Bounan S, Mahdhi A, Drouin D, Ahriz-Saksi S, Guimiot F, Rouas-Freiss N. Immunologic aspects of preeclampsia. AJOG GLOBAL REPORTS 2024; 4:100321. [PMID: 38586611 PMCID: PMC10994979 DOI: 10.1016/j.xagr.2024.100321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024] Open
Abstract
Preeclampsia is a syndrome with multiple etiologies. The diagnosis can be made without proteinuria in the presence of dysfunction of at least 1 organ associated with hypertension. The common pathophysiological pathway includes endothelial cell activation, intravascular inflammation, and syncytiotrophoblast stress. There is evidence to support, among others, immunologic causes of preeclampsia. Unlike defense immunology, reproductive immunology is not based on immunologic recognition systems of self/non-self and missing-self but on immunotolerance and maternal-fetal cellular interactions. The main mechanisms of immune escape from fetal to maternal immunity at the maternal-fetal interface are a reduction in the expression of major histocompatibility complex molecules by trophoblast cells, the presence of complement regulators, increased production of indoleamine 2,3-dioxygenase, activation of regulatory T cells, and an increase in immune checkpoints. These immune protections are more similar to the immune responses observed in tumor biology than in allograft biology. The role of immune and nonimmune decidual cells is critical for the regulation of trophoblast invasion and vascular remodeling of the uterine spiral arteries. Regulatory T cells have been found to play an important role in suppressing the effectiveness of other T cells and contributing to local immunotolerance. Decidual natural killer cells have a cytokine profile that is favored by the presence of HLA-G and HLA-E and contributes to vascular remodeling. Studies on the evolution of mammals show that HLA-E, HLA-G, and HLA-C1/C2, which are expressed by trophoblasts and their cognate receptors on decidual natural killer cells, are necessary for the development of a hemochorial placenta with vascular remodeling. The activation or inhibition of decidual natural killer cells depends on the different possible combinations between killer cell immunoglobulin-like receptors, expressed by uterine natural killer cells, and the HLA-C1/C2 antigens, expressed by trophoblasts. Polarization of decidual macrophages in phenotype 2 and decidualization of stromal cells are also essential for high-quality vascular remodeling. Knowledge of the various immunologic mechanisms required for adequate vascular remodeling and their dysfunction in case of preeclampsia opens new avenues of research to identify novel biological markers or therapeutic targets to predict or prevent the onset of preeclampsia.
Collapse
Affiliation(s)
- Henri Boulanger
- Department of Nephrology and Dialysis, Clinique de l'Estrée, Stains, France (Drs Boulanger and Ahriz-Saksi)
| | - Stéphane Bounan
- Department of Obstetrics and Gynecology, Saint-Denis Hospital Center, Saint-Denis, France (Drs Bounan and Mahdhi)
| | - Amel Mahdhi
- Department of Obstetrics and Gynecology, Saint-Denis Hospital Center, Saint-Denis, France (Drs Bounan and Mahdhi)
| | - Dominique Drouin
- Department of Obstetrics and Gynecology, Clinique de l'Estrée, Stains, France (Dr Drouin)
| | - Salima Ahriz-Saksi
- Department of Nephrology and Dialysis, Clinique de l'Estrée, Stains, France (Drs Boulanger and Ahriz-Saksi)
| | - Fabien Guimiot
- Fetoplacental Unit, Robert-Debré Hospital, Assistance Publique – Hôpitaux de Paris, Paris, France (Dr Guimiot)
| | - Nathalie Rouas-Freiss
- Fundamental Research Division, CEA, Institut de biologie François Jacob, Hemato-Immunology Research Unit, Inserm UMR-S 976, Institut de Recherche Saint-Louis, Paris University, Saint-Louis Hospital, Paris, France (Dr Rouas-Freiss)
| |
Collapse
|
17
|
Chaemsaithong P, Biswas M, Lertrut W, Warintaksa P, Wataganara T, Poon LC, Sukasem C. Pharmacogenomics of Preeclampsia therapies: Current evidence and future challenges for clinical implementation. Best Pract Res Clin Obstet Gynaecol 2024; 92:102437. [PMID: 38103508 DOI: 10.1016/j.bpobgyn.2023.102437] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/03/2023] [Accepted: 11/20/2023] [Indexed: 12/19/2023]
Abstract
Preeclampsia is a pregnancy-specific disorder, and it is a leading cause of maternal and perinatal morbidity and mortality. The application of pharmacogenetics to antihypertensive agents and dose selection in women with preeclampsia is still in its infancy. No current prescribing guidelines from the clinical pharmacogenetics implementation consortium (CPIC) exist for preeclampsia. Although more studies on pharmacogenomics are underway, there is some evidence for the pharmacogenomics of preeclampsia therapies, considering both the pharmacokinetic (PK) and pharmacodynamic (PD) properties of drugs used in preeclampsia. It has been revealed that the CYP2D6*10 variant is significantly higher in women with preeclampsia who are non-responsive to labetalol compared to those who are in the responsive group. Various genetic variants of PD targets, i.e., NOS3, MMP9, MMP2, TIMP1, TIMP3, VEGF, and NAMPT, have been investigated to assess the responsiveness of antihypertensive therapies in preeclampsia management, and they indicated that certain genetic variants of MMP9, TIMP1, and NAMPT are more frequently observed in those who are non-responsive to anti-hypertensive therapies compared to those who are responsive. Further, gene-gene interactions have revealed that NAMPT, TIMP1, and MMP2 genotypes are associated with an increased risk of preeclampsia, and they are more frequently observed in the non-responsive subgroup of women with preeclampsia. The current evidence is not rigorous enough for clinical implementation; however, an institutional or regional-based retrospective analysis of audited data may help close the knowledge gap during the transitional period from a traditional approach (a "one-size-fits-all" strategy) to the pharmacogenomics of preeclampsia therapies.
Collapse
Affiliation(s)
- Piya Chaemsaithong
- Department of Obstetrics and Gynecology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Mohitosh Biswas
- Department of Pharmacy, University of Rajshahi, Rajshahi, Bangladesh; Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand; Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand
| | - Waranyu Lertrut
- Department of Obstetrics and Gynecology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Puntabut Warintaksa
- Department of Obstetrics and Gynecology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Tuangsit Wataganara
- Division of Maternal-Fetal-Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Prannok Road, Bangkoknoi, Bangkok, 10700, Thailand
| | - Liona Cy Poon
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| | - Chonlaphat Sukasem
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand; Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand; Pharmacogenomics Clinic, Bumrungrad Genomic Medicine Institute, Bumrungrad International Hospital, Bangkok, 10110, Thailand; Research and Development Laboratory, Bumrungrad International Hospital, Bangkok, Thailand; Faculty of Pharmaceutical Sciences, Burapha University, Saensuk, Mueang, Chonburi 20131, Thailand; Department of Pharmacology and Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
18
|
Musilova I, Stranik J, Jacobsson B, Kacerovsky M. Antibiotic treatment reduces the intensity of intraamniotic inflammation in pregnancies with idiopathic vaginal bleeding in the second trimester of pregnancy. Am J Obstet Gynecol 2024; 230:245.e1-245.e14. [PMID: 37516399 DOI: 10.1016/j.ajog.2023.07.041] [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: 06/27/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 07/31/2023]
Abstract
BACKGROUND Idiopathic bleeding in the second trimester of pregnancy complicates <1% of all pregnancies. This pregnancy complication can be caused by alterations in local hemostasis in the decidua due to infection/inflammation in the choriodecidual niche. This condition is associated with intraamniotic inflammatory complications. Antibiotic therapy effectively reduces the intensity of intraamniotic inflammation in certain pregnancy pathologies. However, whether antibiotic administration can reduce the intensity of the intraamniotic inflammatory response or eradicate microorganisms in patients with idiopathic bleeding during the second trimester of pregnancy remains unclear. OBJECTIVE This study primarily aimed to determine whether antimicrobial agents can reduce the magnitude of intraamniotic inflammation in patients with idiopathic bleeding in the second trimester of pregnancy by assessing the concentration of interleukin-6 in the amniotic fluid before and after 7 days of antibiotic treatment. The secondary aim was to determine whether treatment with a combination of antibiotics altered the microbial load of Ureaplasma species DNA in amniotic fluid. STUDY DESIGN This retrospective cohort study included singleton-gestation patients with idiopathic bleeding between 15+0 and 27+6 weeks who underwent transabdominal amniocentesis at the time of admission. Follow-up amniocentesis was performed in a subset of patients unless abortion or delivery occurred earlier. Concentrations of interleukin-6 were measured in the amniotic fluid samples, and the presence of microbial invasion of the amniotic cavity was assessed using culture and molecular microbiological methods. Intraamniotic inflammation was defined as an interleukin-6 concentration ≥3000 pg/mL in the amniotic fluid samples. RESULTS A total of 36 patients with idiopathic bleeding in the second trimester of pregnancy were included. All the patients underwent initial amniocentesis. Patients with intraamniotic inflammation (n=25) were treated using a combination of antibiotics consisting of intravenous ceftriaxone, intravenous metronidazole, and peroral clarithromycin. The patients without intraamniotic inflammation (n=11) were treated expectantly. In total, 25 patients delivered 7 days after admission. All patients with intraamniotic inflammation at the initial amniocentesis who delivered after 7 days underwent follow-up amniocentesis. Treatment with antibiotics decreased the interleukin-6 concentration in the amniotic fluid at follow-up amniocentesis compared with that at the initial amniocentesis in patients with intraamniotic inflammation (median [interquartile range]: 3457 pg/mL [2493-13,203] vs 19,812 pg/mL [11,973-34,518]; P=.0001). Amniotic fluid samples with Ureaplasma species DNA had a lower microbial load at the time of follow-up amniocentesis compared with the initial amniocentesis (median [interquartile range]: 1.5×105 copies DNA/mL [1.3×105-1.7×105] vs 8.0×107 copies DNA/mL [6.7×106-1.6×108]; P=.02). CONCLUSION Antibiotic therapy was associated with reduced intraamniotic inflammation in patients with idiopathic bleeding in the second trimester complicated by intraamniotic inflammation. Moreover, antibiotic treatment has been associated with a reduction in the microbial load of Ureaplasma species DNA in the amniotic fluid.
Collapse
Affiliation(s)
- Ivana Musilova
- Biomedical Research Center, University Hospital Hradec Králové, Hradec Králové, Czech Republic; Department of Obstetrics and Gynecology, Hospital Most, Krajská zdravotní a.s., Ústí nad Labem, Czech Republic
| | - Jaroslav Stranik
- Biomedical Research Center, University Hospital Hradec Králové, Hradec Králové, Czech Republic
| | - Bo Jacobsson
- Department of Obstetrics and Gynecology, Institute of Clinical Science, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden; Division of Health Data and Digitalisation, Department of Genetics and Bioinformatics, Norwegian Institute of Public Health, Oslo, Norway
| | - Marian Kacerovsky
- Biomedical Research Center, University Hospital Hradec Králové, Hradec Králové, Czech Republic; Department of Obstetrics and Gynecology, Hospital Most, Krajská zdravotní a.s., Ústí nad Labem, Czech Republic.
| |
Collapse
|
19
|
Jelloul E, Sitoris G, Veltri F, Kleynen P, Rozenberg S, Poppe KG. Gestation-suppressed serum TSH levels during early pregnancy are not associated with altered maternal and neonatal outcomes. Eur Thyroid J 2023; 12:e230112. [PMID: 37855409 PMCID: PMC10620451 DOI: 10.1530/etj-23-0112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/02/2023] [Indexed: 10/04/2023] Open
Abstract
Objective The aim of the study was to investigate the impact of suppressed serum TSH levels (sTSH) during early pregnancy on maternal and neonatal outcomes. Methods In this single-centre, retrospective cohort study 1081 women were screened at 11.8 ± 2.4 weeks of pregnancy for TSH, free T4 (FT4) and TPOAb. Exclusion criteria were twin- and assisted- reproduction pregnancies, women with TSH levels >3.74 mIU/L, severe hyperthyroidism, treated for thyroid dysfunction before or after screening and gestational blood sampling <6 or >16 weeks of pregnancy. The prevalence of adverse pregnancy outcomes was compared between the study group sTSH (TSH: < 0.06 mIU/L; n = 36) and euthyroid controls (TSH: 0.06-3.74 mIU/L; n = 1045), and the impact of sTSH on pregnancy outcomes verified in logistic regression analyses. Results Median (IQR) serum TSH level in women with sTSH was 0.03 (0.03-0.03) vs 1.25 (0.81-1.82) mIU/L in controls and FT4 levels 18.0 (14.4-20.3) vs 14.2 (12.9-15.4) pmol/L; both P < 0.001. None of the women with sTSH had thyrotropin receptor antibodies. Compared with controls, the prevalence of TPOAb positivity (TAI) was comparable between groups (5.6% vs 6.6%; P = 0.803). The prevalence of maternal and neonatal pregnancy outcomes was comparable between the study and control group. The logistic regression analyses with corrections for TAI, FT4 and demographic parameters confirmed the absence of an association between sTSH, and the following outcomes: iron deficient anaemia (aORs (95% CI)): 1.41 (0.64-2.99); P = 0.385, gestational diabetes: 1.19 (0.44-2.88); P = 0.713, preterm birth: 1.57 (0.23-6.22);P = 0.574 and low Apgar-1' score: 0.71 (0.11-2.67); P = 0.657. Conclusions Suppressed serum TSH levels during the first to early second trimester of pregnancy were not associated with altered maternal or neonatal outcomes.
Collapse
Affiliation(s)
- Emna Jelloul
- Endocrine Unit Centre Hospitalier Universitaire (CHU) Saint Pierre, Université Libre de Bruxelles (ULB), Rue Haute, Brussels, Belgium
| | - Georgiana Sitoris
- Endocrine Unit Centre Hospitalier Universitaire (CHU) Saint Pierre, Université Libre de Bruxelles (ULB), Rue Haute, Brussels, Belgium
| | - Flora Veltri
- Endocrine Unit Centre Hospitalier Universitaire (CHU) Saint Pierre, Université Libre de Bruxelles (ULB), Rue Haute, Brussels, Belgium
| | - Pierre Kleynen
- Endocrine Unit Centre Hospitalier Universitaire (CHU) Saint Pierre, Université Libre de Bruxelles (ULB), Rue Haute, Brussels, Belgium
| | - Serge Rozenberg
- Departement of Gynecology and Obstetrics, Centre Hospitalier Universitaire Saint Pierre, Université Libre de Bruxelles (ULB), Rue Haute, Brussels, Belgium
| | - Kris G Poppe
- Endocrine Unit Centre Hospitalier Universitaire (CHU) Saint Pierre, Université Libre de Bruxelles (ULB), Rue Haute, Brussels, Belgium
| |
Collapse
|
20
|
Scioscia M, Siwetz M, Robillard PY, Brizzi A, Huppertz B. Placenta and maternal endothelium during preeclampsia: Disruption of the glycocalyx explains increased inositol phosphoglycans and angiogenic factors in maternal blood. J Reprod Immunol 2023; 160:104161. [PMID: 37857160 DOI: 10.1016/j.jri.2023.104161] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/10/2023] [Accepted: 10/04/2023] [Indexed: 10/21/2023]
Abstract
The etiology of the pregnancy syndrome preeclampsia is still unclear, while most hypotheses center on the placenta as the major contributor of the syndrome. Especially changes of the placental metabolism, including the use of glucose to produce energy, are important features. As an example, inositol phosphoglycan P-type molecules, second messengers involved in the glucose metabolism of all cells, can be retrieved from maternal urine of preeclamptic women, even before the onset of clinical symptoms. Alterations in the placental metabolism may subsequently lead to negative effects on the plasma membrane of the placental syncytiotrophoblast. This in turn may have deleterious effects on the glycocalyx of this layer and a disruption of this layer in all types of preeclampsia. The interruption of the glycocalyx in preeclampsia may result in changes of inositol phosphoglycan P-type signaling pathways and the release of these molecules as well as the release of soluble receptors such as sFlt-1 and sEndoglin. The release of placental factors later affects the maternal endothelium and disrupts the endothelial glycocalyx as well. This in turn may pave the way for edema, endothelial dysfunction, coagulation, all typical symptoms of preeclampsia.
Collapse
Affiliation(s)
- Marco Scioscia
- Department of Obstetrics and Gynecology, Mater Dei Hospital, Via SF Hahnemann 10, 70125 Bari, Italy.
| | - Monika Siwetz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
| | - Pierre-Yves Robillard
- Centre d'Études Périnatales Océan Indien, Centre Hospitalier Universitaire Sud Réunion, Saint-Pierre, La Réunion, France; Service de Néonatologie, Centre Hospitalier Universitaire Sud Réunion, Saint-Pierre, La Réunion, France, Centre Hospitalier Universitaire Sud Réunion, Saint-Pierre, La Réunion, France
| | - Agostino Brizzi
- General and Locoregional Anesthesia Department, Santa Maria Clinic, Via A de Ferrariis, 22, 70124 Bari, Italy
| | - Berthold Huppertz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
| |
Collapse
|
21
|
Admati I, Skarbianskis N, Hochgerner H, Ophir O, Weiner Z, Yagel S, Solt I, Zeisel A. Two distinct molecular faces of preeclampsia revealed by single-cell transcriptomics. MED 2023; 4:687-709.e7. [PMID: 37572658 DOI: 10.1016/j.medj.2023.07.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/04/2023] [Accepted: 07/14/2023] [Indexed: 08/14/2023]
Abstract
INTRODUCTION Preeclampsia is a multisystemic, pregnancy-specific disorder united by new-onset hypertension but with considerable variation in clinical manifestation, onset, and severity. For symptoms to regress, delivery of the placenta is required. For symptoms to regress, delivery of the placenta is required, making the placenta central to preeclampsia pathophysiology. To dissect which placental functions were impacted in two forms of preeclampsia, we studied molecular changes across the cell types of the placenta. METHODS We performed a transcriptomic survey of single-cells and single-nuclei on cases of early- and late-onset preeclampsia with gestation-matched controls. FINDINGS Our data revealed massive dysregulation of gene expression in all cell classes that was almost exclusive to early preeclampsia. For example, an important known receptor/ligand imbalance hallmarking angiogenic disfunction, sFLT1/placental growth factor (PGF), was reflected in striking, cell-autonomous dysregulation of FLT1 and PGF transcription in the syncytium in early preeclampsia only. Stromal cells and vasculature echoed an inflamed, stressed, anti-angiogenic environment. Finally, the placental immune niche set the tone for inflammation in early but not late preeclampsia. Here, fetal-origin Hofbauer and maternal-origin TREM2 macrophages were revealed as surprising main actors, while local cells of the adaptive immune system were largely unaffected. Late preeclampsia showed minimal cellular impact on the placenta. CONCLUSIONS Our survey provides systematic molecular evidence for two distinct diseases. We resolved systematic molecular dysregulation to individual cell types with strong implications for definition, early detection, diagnosis, and treatment. FUNDING Funded by the Preeclampsia Foundation through the Peter Joseph Pappas Research Grant.
Collapse
Affiliation(s)
- Inbal Admati
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Niv Skarbianskis
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Hannah Hochgerner
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Osnat Ophir
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Zeev Weiner
- Department of Obstetrics and Gynecology, Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Simcha Yagel
- Division of Obstetrics and Gynecology Hadassah, Hebrew University Medical Centers, Jerusalem, Israel
| | - Ido Solt
- Department of Obstetrics and Gynecology, Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel.
| | - Amit Zeisel
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
22
|
Premkumar A, Rana S. Reducing the Risk of Term Preeclampsia by Risk-Stratified Timing of Birth: Time to Deliver. Hypertension 2023; 80:979-980. [PMID: 37075132 DOI: 10.1161/hypertensionaha.123.20972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Affiliation(s)
- Ashish Premkumar
- Section of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Pritzker School of Medicine, University of Chicago, IL (A.P., S.R.)
- Department of Anthropology, The Graduate School, Northwestern University, Evanston, IL (A.P.)
| | - Sarosh Rana
- Section of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Pritzker School of Medicine, University of Chicago, IL (A.P., S.R.)
| |
Collapse
|
23
|
Chaiworapongsa T, Romero R, Gotsch F, Gomez-Lopez N, Suksai M, Gallo DM, Jung E, Levenson D, Tarca AL. One-third of patients with eclampsia at term do not have an abnormal angiogenic profile. J Perinat Med 2022:jpm-2022-0474. [PMID: 36567427 DOI: 10.1515/jpm-2022-0474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/04/2022] [Indexed: 12/27/2022]
Abstract
OBJECTIVES An abnormal angiogenic profile is present in about one-half of women with preeclampsia at term. Few studies examined the roles of angiogenic biomarkers in eclampsia. The aims of this study were to determine (1) whether the degree of an anti-angiogenic state, reflected by a low placental growth factor (PlGF) to soluble fms-like tyrosine kinase-1 (sFlt-1) ratio, in women with eclampsia differed from that of women with severe preeclampsia; and (2) the prevalence of women who had an abnormal angiogenic profile at the diagnoses of preterm and term eclampsia. METHODS A cross-sectional study was conducted to include women in the following groups: (1) uncomplicated pregnancy (n=40); (2) severe preeclampsia (n=50); and (3) eclampsia (n=35). Maternal serum concentrations of PlGF and sFlt-1 were determined by immunoassays. RESULTS Women with preterm, but not term, eclampsia had a more severe anti-angiogenic state than those with severe preeclampsia (lower PlGF and PlGF/sFlt-1 ratio, each p<0.05). However, the difference diminished in magnitude with increasing gestational age (interaction, p=0.005). An abnormal angiogenic profile was present in 95% (19/20) of women with preterm eclampsia but in only 67% (10/15) of women with eclampsia at term. CONCLUSIONS Angiogenic biomarkers can be used for risk assessment of preterm eclampsia. By contrast, a normal profile of angiogenic biomarkers cannot reliably exclude patients at risk for eclampsia at term. This observation has major clinical implications given that angiogenic biomarkers are frequently used in the triage area as a test to rule out preeclampsia.
Collapse
Affiliation(s)
- Tinnakorn Chaiworapongsa
- 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, U. S. Department of Health and Human Services, National Institutes of Health, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, 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, U. S. Department of Health and Human Services, National Institutes of Health, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA.,Detroit Medical Center, Detroit, MI, USA
| | - Francesca Gotsch
- 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, U. S. Department of Health and Human Services, National Institutes of Health, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, 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, U. S. Department of Health and Human Services, National Institutes of Health, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Manaphat Suksai
- 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, U. S. Department of Health and Human Services, National Institutes of Health, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Dahiana M Gallo
- 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, U. S. Department of Health and Human Services, National Institutes of Health, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Obstetrics and Gynecology, University of Valle, Cali, Colombia
| | - Eunjung Jung
- 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, U. S. Department of Health and Human Services, National Institutes of Health, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Dustyn Levenson
- 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, U. S. Department of Health and Human Services, National Institutes of Health, Bethesda, MD, and Detroit, MI, USA.,Wayne State University School of Medicine, Detroit, MI, 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, U. S. Department of Health and Human Services, National Institutes of Health, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA
| |
Collapse
|