1
|
Vincent KMM, Garner T, Stevens A, Cottrell EC, Myers JE, Higgins LE. Assessing Placental Dysfunction Subtypes in Pregnancies With a Low PlGF Centile. Hypertension 2025. [PMID: 40396254 DOI: 10.1161/hypertensionaha.124.24440] [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: 12/04/2024] [Accepted: 04/29/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND A low circulating placental growth factor (PlGF) concentration is considered a marker of placental dysfunction, the predominant cause of preeclampsia and fetal growth restriction. Besides iatrogenic delivery, there are no effective treatments, potentially due to its heterogeneity. We hypothesize that >1 subtype of placental dysfunction exists in pregnancies with a low circulating PlGF. METHODS Matched placental villous and maternal plasma samples (low PlGF <5th, n=19; normal PlGF ≥5th centile, n=20) were collected from uncomplicated pregnancies and those complicated by 1 or a combination of preeclampsia, fetal growth restriction, or chronic hypertension. Transcriptomic and metabolomic data sets were obtained from villous samples and used to perform cluster-of-cluster analysis. Clinical outcomes were compared between clusters, as well as subsequent pathway analysis. RESULTS Multiomic cluster-of-cluster analysis resulted in 3 molecular clusters. Cluster 1 predominantly consisted of those with a low PlGF (9/10); PlGF results in cluster 2 varied (low, n=8/20; normal, n=12/20), while cluster 3 mainly comprised of those with a normal PlGF result (n=7/9). Birthweight was significantly lower in cluster 1, as well as an increased incidence of early-onset preeclampsia. However, pathways commonly associated with placental dysfunction were only identified in cluster 2. CONCLUSIONS Multiomic analysis revealed 2 potential subtypes of placental dysfunction associated with a low circulating PlGF. These results support previous studies suggesting the existence of preeclampsia subtypes. Clinical outcome comparisons indicate cluster 1 as the severe subtype; however, pathway analysis contradicted this. Improved understanding of subtype etiology could enable a more personalized therapeutic approach for pregnancies complicated by placental dysfunction.
Collapse
Affiliation(s)
- Kirsty M M Vincent
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, United Kingdom (K.M.M.V., T.G., A.S., E.C.C., J.E.M., L.E.H.)
| | - Terence Garner
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, United Kingdom (K.M.M.V., T.G., A.S., E.C.C., J.E.M., L.E.H.)
| | - Adam Stevens
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, United Kingdom (K.M.M.V., T.G., A.S., E.C.C., J.E.M., L.E.H.)
| | - Elizabeth C Cottrell
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, United Kingdom (K.M.M.V., T.G., A.S., E.C.C., J.E.M., L.E.H.)
| | - Jenny E Myers
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, United Kingdom (K.M.M.V., T.G., A.S., E.C.C., J.E.M., L.E.H.)
- Manchester University Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, United Kingdom (J.E.M., L.E.H.)
| | - Lucy E Higgins
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, United Kingdom (K.M.M.V., T.G., A.S., E.C.C., J.E.M., L.E.H.)
- Manchester University Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, United Kingdom (J.E.M., L.E.H.)
| |
Collapse
|
2
|
Starodubtseva N, Poluektova A, Tokareva A, Kukaev E, Avdeeva A, Rimskaya E, Khodzayeva Z. Proteome-Based Maternal Plasma and Serum Biomarkers for Preeclampsia: A Systematic Review and Meta-Analysis. Life (Basel) 2025; 15:776. [PMID: 40430203 PMCID: PMC12113278 DOI: 10.3390/life15050776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 04/29/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Proteomics has emerged as a transformative tool in biomedical research, enabling comprehensive characterization of protein profiles in complex biological systems. In preeclampsia (PE) research, quantitative proteomic analyses of plasma and serum have revealed critical insights into disease mechanisms and potential biomarkers. Through a systematic review of 17 studies (2009-2024), we identified 561 differentially expressed plasma/serum proteins (p < 0.05) in PE patients versus healthy controls, with 122 proteins consistently replicated across ≥2 independent studies. Stratified analysis by clinical subtype (early-vs. late-onset PE) demonstrated both concordant and divergent protein expression patterns, reflecting heterogeneity in PE pathophysiology, methodological variations (e.g., sample processing, proteomic platforms), and differences between discovery-phase and targeted validation studies. The trimester-specific biomarker panels proposed here offer a framework for future large-scale, multicenter validation. By integrating advanced proteomic technologies with standardized preanalytical and analytical protocols, these findings advance opportunities for early prediction (first-trimester biomarker signatures); mechanistic insight (complement system involvement); and personalized management (subtype-specific therapeutic targets). This work underscores the potential of proteomics to reshape PE research, from molecular discovery to clinical translation, ultimately improving outcomes for this leading cause of maternal and perinatal morbidity.
Collapse
Affiliation(s)
- Natalia Starodubtseva
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
| | - Alina Poluektova
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
| | - Alisa Tokareva
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
| | - Evgenii Kukaev
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
- V.L. Talrose Institute for Energy Problems of Chemical Physics, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Anna Avdeeva
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
| | - Elena Rimskaya
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
- Lebedev Physical Institute, 119991 Moscow, Russia
| | - Zulfiya Khodzayeva
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
| |
Collapse
|
3
|
Taylor BD, Haggerty CL, Amabebe E, Richardson LS. Current Evidence of Maternal Infection With Chlamydia trachomatis and Preeclampsia Risk. Am J Reprod Immunol 2025; 93:e70080. [PMID: 40298141 DOI: 10.1111/aji.70080] [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: 07/19/2024] [Revised: 01/13/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
Chlamydia trachomatis is the most common bacterial sexually transmitted infection (STI) in the United States. Ascending C. trachomatis can cause pelvic inflammatory disease (PID), potentially leading to subsequent infertility, ectopic pregnancy, and adverse pregnancy outcomes. There is growing evidence implicating infections (e.g., COVID-19, cytomegalovirus) in preeclampsia etiology, a maternal hypertensive disorder and leading cause of maternal morbidity and mortality. However, few studies have investigated the impact of STIs on preeclampsia risk. In this review, we provide an overview of the potential association between C. trachomatis and preeclampsia and identify future research needs through a critical evaluation of epidemiologic, in vitro, and in vivo studies. Unfortunately, current methodological limitations such as lower-quality study designs, selection bias, confounding bias, and variations in chlamydia diagnostic methods inhibit our understanding of the impact of C. trachomatis on preeclampsia. In addition, bench-side approaches such as animal models and in vitro studies have not elucidated the mechanisms linking C. trachomatis to preeclampsia. Understanding the biological pathways that could be disrupted by chlamydia is important as it may ultimately guide the development and use of novel therapeutics to augment standard antibiotic therapy to reduce pathology.
Collapse
Affiliation(s)
- Brandie DePaoli Taylor
- Department of Obstetrics and Gynecology, Division of Basic Science and Translational Research, University of Texas Medical Branch, Galveston, Texas, USA
- Academic Research, Advocate Aurora Research Institute, Milwaukee, Wisconsin, USA
| | - Catherine L Haggerty
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Emmanuel Amabebe
- Department of Obstetrics and Gynecology, Division of Basic Science and Translational Research, University of Texas Medical Branch, Galveston, Texas, USA
| | - Lauren S Richardson
- Department of Obstetrics and Gynecology, Division of Basic Science and Translational Research, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
4
|
Keuls RA, Ochsner SA, O'Neill MB, O'Day DR, Miyauchi A, Campbell KM, Lanners N, Goldstein JA, Yee C, McKenna NJ, Parchem RJ, Parchem JG. Single-nucleus transcriptional profiling of the placenta reveals the syncytiotrophoblast stress response to COVID-19. Am J Obstet Gynecol 2025; 232:S160-S175.e7. [PMID: 40253079 DOI: 10.1016/j.ajog.2025.01.028] [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: 07/22/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND COVID-19 in pregnancy is associated with placental immune activation, inflammation, and vascular malperfusion, but its impact on syncytiotrophoblast biology and function is unclear. OBJECTIVE This study aimed to determine the effects of maternal COVID-19 on placental syncytiotrophoblasts using single-nucleus transcriptional profiling and to compare placental stress responses in COVID-19 and preeclampsia. STUDY DESIGN For transcriptional characterization of syncytiotrophoblasts, we used the single-nucleus RNA sequencing platform, single-cell combinatorial indexing RNA sequencing (sci-RNA-seq3), to profile placental villi and fetal membranes from unvaccinated patients with symptomatic COVID-19 at birth (n = 4), gestational age-matched controls (n = 4), and a case of critical COVID-19 in the second trimester with delivery at term (n = 1). Clustering of nuclei and differential gene expression analysis was performed in Seurat. Gene ontology analysis was conducted using Enrichr. High-confidence transcriptional target analysis was used to identify key transcription factor nodes governing the syncytiotrophoblast response to maternal SARS-CoV-2 infection. Bioinformatic approaches were further used to compare the COVID-19 dataset to published preeclampsia gene signatures. Tissue analysis, including immunofluorescence, was conducted to validate the transcriptional data and to compare COVID-19 and preeclampsia placental histology for an expanded cohort of placentas: controls (n = 6), asymptomatic COVID-19 (n = 3), symptomatic COVID-19 (n = 5), and preeclampsia with severe features (n = 7). RESULTS The analyzed dataset comprised 15 cell clusters and 47,889 nuclei. We identified 3 clusters of syncytiotrophoblasts representing fusing and mature nuclei with overlapping but distinct transcriptional responses to COVID-19. Bioinformatic analyses indicated that COVID-19 is associated with the following alterations in syncytiotrophoblasts: (1) endoplasmic reticulum stress and activation of stress signaling pathways, including the unfolded protein response and integrated stress response; (2) regulation of gene expression by CCAAT/enhancer-binding protein beta (CEBPB), a master transcription factor of the syncytiotrophoblast lineage; and (3) upregulation of preeclampsia-associated genes. Using complementary methods, we confirmed increased levels of stress response proteins (eg, BiP, G3BP1) in syncytiotrophoblasts, unfolded protein response signaling (spliced XBP1 mRNA), and CEBPB activation (phosphorylation) in COVID-19. Increased cytotrophoblast proliferation (Ki-67) was also detected in COVID-19, consistent with a trophoblast response to injury. Markers of stress detected in preeclampsia demonstrated similarities in the placental stress phenotype of COVID-19 and preeclampsia. CONCLUSION Maternal COVID-19 is associated with syncytiotrophoblast endoplasmic reticulum stress and activation of the syncytiotrophoblast lineage transcription factor, CEBPB. Similarities between syncytiotrophoblast stress in COVID-19 and preeclampsia provide insights into their clinical association.
Collapse
Affiliation(s)
- Rachel A Keuls
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX; Department of Neuroscience, Baylor College of Medicine, Houston, TX
| | - Scott A Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Mary B O'Neill
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA
| | - Diana R O'Day
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA
| | - Akihiko Miyauchi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX; Department of Neuroscience, Baylor College of Medicine, Houston, TX
| | - Kadeshia M Campbell
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX
| | - Natalie Lanners
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX
| | - Jeffery A Goldstein
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Connor Yee
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX; Larry C. Gilstrap MD Center for Perinatal and Women's Health Research, The University of Texas Health Science Center at Houston, Houston, TX
| | - Neil J McKenna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Ronald J Parchem
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX; Department of Neuroscience, Baylor College of Medicine, Houston, TX.
| | - Jacqueline G Parchem
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX.
| |
Collapse
|
5
|
Pan H, Ouyang B, Zhang H, Zhao C. Non-coding RNAs: the architects of placental development and pregnancy success. Mol Genet Genomics 2025; 300:39. [PMID: 40159439 DOI: 10.1007/s00438-025-02244-8] [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: 01/10/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025]
Abstract
Noncoding RNAs (ncRNAs) constitute a significant portion of the transcriptome that lacks evident protein-coding functions; however, they have been confirmed to be crucial in various biological processes, including placental development. Notwithstanding the existence of various ncRNAs, research on their role in placental development and pregnancy has been constrained. The predominant category of identified ncRNAs specific to placental tissue is microRNAs (miRNAs). Given their prevalence, the significantly larger cohort of other non-coding RNAs, such as circular RNAs (circRNAs) and long non-coding RNAs (lncRNAs), is anticipated to exert a considerably greater influence than miRNAs. Syncytiotrophoblast, a fetal-derived cell, serves as a conduit between the fetus and mother by secreting extracellular vesicles that contain fetal proteins and RNA. Alterations in ncRNAs within placental tissue, especially in trophoblast cells and extracellular vesicles, may be linked to placental dysfunction that leads to pregnancy complications, serving either as a causative factor or a result. This review encapsulates the existing understanding of ncRNAs in placental development, pregnancy success, pregnancy-related complications, extracellular vesicle conveyance, and their capacity as innovative diagnostic instruments and therapeutic strategies.
Collapse
Affiliation(s)
- Hongjuan Pan
- Taikang Tongji (Wuhan) Hospital, Wuhan, 430050, Hubei, China
| | - Baisha Ouyang
- Taikang Tongji (Wuhan) Hospital, Wuhan, 430050, Hubei, China
| | - Hui Zhang
- Taikang Tongji (Wuhan) Hospital, Wuhan, 430050, Hubei, China
| | - Caizhen Zhao
- Taikang Tongji (Wuhan) Hospital, Wuhan, 430050, Hubei, China.
| |
Collapse
|
6
|
Alhasan MM, Landa MS, García SI, Gerlach RG, Harb H, Fahlbusch FB, Conrad ML, Barrientos G. Distinct gut microbial signature and altered short chain fatty acid metabolism at disease onset in a rat preclinical model of superimposed preeclampsia. Sci Rep 2024; 14:32137. [PMID: 39738527 PMCID: PMC11686345 DOI: 10.1038/s41598-024-83981-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/18/2024] [Indexed: 01/02/2025] Open
Abstract
Chronic hypertension is an increasingly prevalent condition that constitutes a risk factor for superimposed preeclampsia during pregnancy. In this study, we assessed the gut microbiome in a rat model of superimposed preeclampsia to characterize the microbial signature associated with defective placentation processes identified at the preclinical disease stage. The blood pressure profile, renal function parameters and fetal phenotype were evaluated in pregnant Stroke-prone Spontaneously Hypertensive Rats (SHRSP) and their normotensive controls. On gestation day (GD)14, feces were collected and gut microbiome composition and short-chain fatty acid concentrations were determined by 16S rRNA sequencing and gas chromatography respectively. At disease onset on GD14, the fecal gut microbiome of SHRSP showed a lower alpha diversity and significant differences in beta diversity when compared with control animals. In the feces, Prevotella, Bifidobacterium, Parasutterella and Roseburia were enriched in SHRSP pregnancies compared to controls, showing a strong correlation with clinical parameters. Bacteria from the families Ruminococcaceae, Oscillospiraceae and the genera Blautia and Faecalibacterium were depleted. Considering short-chain fatty acids, acetate, propionate and valerate were increased in the SHRSP model, showing a strong positive correlation with the relative abundance of enriched taxa. We show that on GD14, at the asymptomatic SPE onset stage, pregnant SHRSP display a distinct gut microbiome signature and altered short chain fatty acid metabolism compared to control animals.
Collapse
Affiliation(s)
- Moumen M Alhasan
- Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - María S Landa
- Facultad de Medicina, Instituto de Investigaciones Médicas Alfredo Lanari, Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires, Argentina
- Laboratorio de Cardiología Molecular, Instituto de Investigaciones Médicas (IDIM), Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Silvia I García
- Facultad de Medicina, Instituto de Investigaciones Médicas Alfredo Lanari, Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires, Argentina
- Laboratorio de Cardiología Molecular, Instituto de Investigaciones Médicas (IDIM), Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
- Laboratorio de Medicina Experimental, Hospital Alemán, Av. Pueyrredón 1640, C1118AAT, Ciudad Autónoma de Buenos Aires, Argentina
| | - Roman G Gerlach
- Institute of Clinical Microbiology, Immunology and Hygiene, University Hospital of Erlangen and Friedrich-Alexander-University (FAU) Erlangen-Nuremberg, Erlangen, Germany
| | - Hani Harb
- Institute for Medical Microbiology and Virology, Medical Faculty, TU Dresden, Dresden, Germany
| | - Fabian B Fahlbusch
- Neonatology and Pediatric Intensive Care, Faculty of Medicine, University of Augsburg, 86156, Augsburg, Germany
| | - Melanie L Conrad
- Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health, Berlin, Germany
- Neonatology and Pediatric Intensive Care, Faculty of Medicine, University of Augsburg, 86156, Augsburg, Germany
| | - Gabriela Barrientos
- Laboratorio de Medicina Experimental, Hospital Alemán, Av. Pueyrredón 1640, C1118AAT, Ciudad Autónoma de Buenos Aires, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
7
|
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 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] [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, 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, 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, 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
|
8
|
Maher GM, Kenny LC, Navaratnam K, Alfirevic Z, Sheehan D, Baker PN, Gluud C, Tuytten R, Kublickas M, Niklasson B, Duvekot JJ, van den Berg CB, Wu P, Kublickiene K, McCarthy FP, Khashan AS. Cohort profile: Improved Pregnancy Outcomes via Early Detection (IMPROvED), an International Multicentre Prospective Cohort. HRB Open Res 2024; 6:65. [PMID: 38911611 PMCID: PMC11190647 DOI: 10.12688/hrbopenres.13812.2] [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: 06/21/2024] [Indexed: 06/25/2024] Open
Abstract
Background Improved Pregnancy Outcomes via Early Detection (IMPROvED) is a multi-centre, European phase IIa clinical study. The primary aim of IMPROvED is to enable the assessment and refinement of innovative prototype preeclampsia risk assessment tests based on emerging biomarker technologies. Here we describe IMPROvED's profile and invite researchers to collaborate. Methods A total of 4,038 low-risk nulliparous singleton pregnancies were recruited from maternity units in Ireland (N=1,501), United Kingdom (N=1,108), The Netherlands (N=810), and Sweden (N=619) between November 2013 to August 2017. Participants were interviewed by a research midwife at ~11 weeks (optional visit), ~15 weeks, ~20 weeks, ~34 weeks' gestation (optional visit), and postpartum (within 72-hours following delivery). Findings to date Clinical data included information on maternal sociodemographic, medical history, and lifestyle factors collected at ~15 weeks' gestation, and maternal measurements, collected at each study visit. Biobank samples included blood, urine, and hair collected at each study visit throughout pregnancy in all units plus umbilical cord/blood samples collected at birth in Ireland and Sweden. A total of 74.0% (N=2,922) had an uncomplicated pregnancy, 3.1% (N=122) developed preeclampsia, 3.6% (N=143) had a spontaneous preterm birth, and 10.5% (N=416) had a small for gestational age baby. We evaluated a panel of metabolite biomarkers and a panel of protein biomarkers at 15 weeks and 20 weeks' gestation for preeclampsia risk assessment. Their translation into tests with clinical application, as conducted by commercial entities, was hampered by technical issues and changes in test requirements. Work on the panel of proteins was abandoned, while work on the use of metabolite biomarkers for preeclampsia risk assessment is ongoing. Future plans In accordance with the original goals of the IMPROvED study, the data and biobank are now available for international collaboration to conduct high quality research into the cause and prevention of adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Gillian M. Maher
- INFANT Research Centre, University College Cork, Cork, T12YE02, Ireland
- School of Public Health, University College Cork, Cork, T12XF62, Ireland
| | - Louise C. Kenny
- Department of Women’s and Children’s Health, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, L693BX, UK
| | - Kate Navaratnam
- Department of Women’s and Children’s Health, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, L693BX, UK
| | - Zarko Alfirevic
- Department of Women’s and Children’s Health, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, L693BX, UK
| | - Darina Sheehan
- INFANT Research Centre, University College Cork, Cork, T12YE02, Ireland
| | - Philip N. Baker
- College of Life Sciences, University of Leicester, Leicester, LE17RH, UK
| | - Christian Gluud
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Copenhagen University Hospital - Rigshospitalet, Copenhagen, The Capital Region, Copenhagen, DK2200, Denmark
- Department of Regional Health Research, The Faculty of Health Sciences, University of Southern Denmark, Odense, DK5230, Denmark
| | | | - Marius Kublickas
- Department of Fetal Medicine, Karolinska University Hospital, Stockholm, SE17176, Sweden
| | - Boel Niklasson
- Department of Nursing Science, Sophiahemmet University, Stockholm, SE11486, Sweden
| | - Johannes J. Duvekot
- Department of Obstetrics and Gynecology, Division of Obstetrics and Prenatal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, 3015GD, The Netherlands
| | - Caroline B. van den Berg
- Department of Obstetrics and Gynecology, Division of Obstetrics and Prenatal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, 3015GD, The Netherlands
| | - Pensee Wu
- School of Medicine, Keele University, Staffordshire, ST55BG, UK
| | - Karolina Kublickiene
- Division of Renal Medicine, CLINTEC, Karolinska Institutet, Stockholm, SE14152, Sweden
| | - Fergus P. McCarthy
- INFANT Research Centre, University College Cork, Cork, T12YE02, Ireland
- Department of Obstetrics and Gynaecology, University College Cork, Cork, T12YE02, Ireland
| | - Ali S. Khashan
- INFANT Research Centre, University College Cork, Cork, T12YE02, Ireland
- School of Public Health, University College Cork, Cork, T12XF62, Ireland
| |
Collapse
|
9
|
Balduit A, Agostinis C, Mangogna A, Zito G, Stampalija T, Ricci G, Bulla R. Systematic review of the complement components as potential biomarkers of pre-eclampsia: pitfalls and opportunities. Front Immunol 2024; 15:1419540. [PMID: 38983853 PMCID: PMC11232388 DOI: 10.3389/fimmu.2024.1419540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/07/2024] [Indexed: 07/11/2024] Open
Abstract
The complement system (C) is a crucial component of the innate immune system. An increasing body of research has progressively shed light on the pivotal role of C in immunological tolerance at the feto-maternal interface. Excessive C activation or impaired C regulation may determine the onset of pregnancy-related pathological conditions, including pre-eclampsia (PE). Thus, several studies have investigated the presence of C components or split products in blood matrixes (i.e., plasma, serum), urine, and amniotic fluid in PE. In the current study, we systematically reviewed the currently available scientific literature reporting measurements of C components as circulating biomarkers in PE, based on a literature search using Pubmed, Scopus, and Embase databases. A total of 41 out of 456 studies were selected after full-text analysis. Fourteen studies (34.1%) were identified as measuring the blood concentrations of the classical pathway, 5 (12.1%) for the lectin pathway, 28 (68.3%) for the alternative pathway, 17 (41.5%) for the terminal pathway components, and 16 (39%) for C regulators. Retrieved results consistently reported C4, C3, and factor H reduction, and increased circulating levels of C4d, Bb, factor D, C3a, C5a, and C5b-9 in PE compared to normal pregnancies, depicting an overall scenario of excessive C activation and aberrant C regulation. With evidence of C activation and dysregulation, C-targeted therapy is an intriguing perspective in PE management. Moreover, we also discussed emerging pitfalls in C analysis, mainly due to a lack of experimental uniformity and biased cohort selection among different studies and laboratories, aiming to raise a more comprehensive awareness for future standardization. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024503070.
Collapse
Affiliation(s)
- Andrea Balduit
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Chiara Agostinis
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Alessandro Mangogna
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Gabriella Zito
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Tamara Stampalija
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
- Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Giuseppe Ricci
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
- Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
10
|
Barrientos G, Schuman ML, Landa MS, Robello E, Incardona C, Conrad ML, Galleano M, García SI. Therapeutic Effect of Alpha Lipoic Acid in a Rat Preclinical Model of Preeclampsia: Focus on Maternal Signs, Fetal Growth and Placental Function. Antioxidants (Basel) 2024; 13:730. [PMID: 38929169 PMCID: PMC11200649 DOI: 10.3390/antiox13060730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/08/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Chronic hypertension is a major risk factor for preeclampsia (PE), associated with significant maternal and neonatal morbidity. We previously demonstrated that pregnant stroke-prone spontaneously hypertensive rats (SHRSP) display a spontaneous PE-like phenotype with distinct placental, fetal, and maternal features. Here, we hypothesized that supplementation with alpha lipoic acid (ALA), a potent antioxidant, during early pregnancy could ameliorate the PE phenotype in this model. To test this hypothesis, timed pregnancies were established using 10 to 12-week-old SHRSP females (n = 19-16/group), which were assigned to two treatment groups: ALA (injected intraperitoneally with 25 mg/kg body weight ALA on gestation day (GD1, GD8, and GD12) or control, receiving saline following the same protocol. Our analysis of maternal signs showed that ALA prevented the pregnancy-dependent maternal blood pressure rise (GD14 blood pressure control 169.3 ± 19.4 mmHg vs. 146.1 ± 13.4 mmHg, p = 0.0001) and ameliorated renal function, as noted by the increased creatinine clearance and improved glomerular histology in treated dams. Treatment also improved the fetal growth restriction (FGR) phenotype, leading to increased fetal weights (ALA 2.19 ± 0.5 g vs. control 1.98 ± 0.3 g, p = 0.0074) and decreased cephalization indexes, indicating a more symmetric fetal growth pattern. This was associated with improved placental efficiency, decreased oxidative stress marker expression on GD14, and serum soluble fms-like tyrosine kinase 1 (sFlt1) levels on GD20. In conclusion, ALA supplementation mitigated maternal signs and improved placental function and fetal growth in SHRSP pregnancies, emerging as a promising therapy in pregnancies at high risk for PE.
Collapse
Affiliation(s)
- Gabriela Barrientos
- Laboratorio de Medicina Experimental, Hospital Alemán, Ciudad Autónoma de Buenos Aires C1118AAT, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires C1118AAT, Argentina
| | - Mariano L. Schuman
- Facultad de Medicina, Instituto de Investigaciones Médicas Alfredo Lanari, Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1053ABH, Argentina; (M.L.S.); (M.S.L.)
- Departamento de Cardiología Molecular, Instituto de Investigaciones Médicas (IDIM), UBA-CONICET, Ciudad Autónoma de Buenos Aires C1427ARN, Argentina
| | - Maria S. Landa
- Facultad de Medicina, Instituto de Investigaciones Médicas Alfredo Lanari, Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1053ABH, Argentina; (M.L.S.); (M.S.L.)
- Departamento de Cardiología Molecular, Instituto de Investigaciones Médicas (IDIM), UBA-CONICET, Ciudad Autónoma de Buenos Aires C1427ARN, Argentina
| | - Elizabeth Robello
- Facultad de Farmacia y Bioquímica, Cátedra de Fisicoquímica, Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1053ABH, Argentina; (E.R.); (M.G.)
- Instituto de Bioquímica y Medicina Molecular-Dr. Alberto Boveris (IBIMOL), UBA-CONICET, Ciudad Autónoma de Buenos Aires C1113AAD, Argentina
| | - Claudio Incardona
- Fundación GADOR, Ciudad Autónoma de Buenos Aires C1414CUI, Argentina;
| | - Melanie L. Conrad
- Institute of Microbiology, Infectious Diseases and Immunology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 12203 Berlin, Germany;
| | - Monica Galleano
- Facultad de Farmacia y Bioquímica, Cátedra de Fisicoquímica, Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1053ABH, Argentina; (E.R.); (M.G.)
- Instituto de Bioquímica y Medicina Molecular-Dr. Alberto Boveris (IBIMOL), UBA-CONICET, Ciudad Autónoma de Buenos Aires C1113AAD, Argentina
| | - Silvia I. García
- Laboratorio de Medicina Experimental, Hospital Alemán, Ciudad Autónoma de Buenos Aires C1118AAT, Argentina;
- Facultad de Medicina, Instituto de Investigaciones Médicas Alfredo Lanari, Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1053ABH, Argentina; (M.L.S.); (M.S.L.)
- Departamento de Cardiología Molecular, Instituto de Investigaciones Médicas (IDIM), UBA-CONICET, Ciudad Autónoma de Buenos Aires C1427ARN, Argentina
| |
Collapse
|
11
|
Rhein AK, Rabinovich A, Abuhasira R, Lubaton-Barshishat S, Erez O. Obstetric antiphospholipid syndrome carries an increased lifetime risk for obstetric and thrombotic complications-a population-based study. Res Pract Thromb Haemost 2024; 8:102430. [PMID: 38798792 PMCID: PMC11127162 DOI: 10.1016/j.rpth.2024.102430] [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] [Received: 02/15/2024] [Revised: 04/13/2024] [Accepted: 04/23/2024] [Indexed: 05/29/2024] Open
Abstract
Background Antiphospholipid syndrome (APS) can present with either a thromboembolic event (thrombotic APS, TAPS) or an obstetric complication (obstetric APS, OAPS). Data on long-term complications in the different APS phenotypes are limited. Objectives We aimed to compare obstetric history, antiphospholipid antibody profiles, obstetric and thromboembolic complications, and pregnancy outcomes between TAPS and OAPS. Methods This retrospective cohort study included women who delivered singleton pregnancies between 1998 and 2020. One hundred sixteen thousand four hundred nine women were included, resulting in 320,455 deliveries. Among the included patients, 71 were diagnosed with APS, 49 were classified as OAPS, and 22 as TAPS. The demographics, obstetric, neonatal, and thrombotic outcomes were compared among TAPS, OAPS, and the general obstetric population. Results OAPS patients had an increased risk of thrombotic events compared with the general obstetric population (odds ratio [OR] 18.0; 95% CI, 8.7-37.2). In pregnancies following the diagnosis of APS, despite standard antithrombotic treatment, OAPS patients exhibited an elevated risk of placenta-related and neonatal complications compared with the general obstetric population (late fetal loss [adjusted OR {aOR}, 15.3; 95% CI, 0.5-27.5], stillbirth [aOR, 5.9; 95% CI, 2.2-15.4], placental abruption [aOR, 4.8; 95% CI, 1.5-15.3], preeclampsia [aOR, 4.4; 95% CI, 2.5-7.7], fetal growth restriction [aOR, 4.3; 95% CI, 8.5-27.5], small for gestational age neonate [aOR, 4.0; 95% CI, 2.4-6.6], and low Apgar scores [Apgar'1: aOR, 2.6; 95% CI, 1.3-10.4; Apgar'5: aOR, 3.7; 95% CI, 1.3-10.4]). TAPS patients exhibited increased risk of preeclampsia (aOR, 3.1; 95% CI, 1.2-8). Conclusion OAPS patients exhibit a heightened risk of thrombotic events compared with the general obstetric population. Despite treatment, OAPS and TAPS still presented obstetric complications. These findings, after confirmation in prospective studies, need to be taken into consideration when planning the treatment approach for these patients.
Collapse
Affiliation(s)
- Ariel Katherine Rhein
- The Joyce & Irving Goldman Medical School, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Anat Rabinovich
- Thrombosis and Hemostasis Unit, Hematology Institute, Soroka University Medical Center and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ran Abuhasira
- The Joyce & Irving Goldman Medical School, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Clinical Research Center, Soroka University Medical Center, Beer-Sheva, Israel
| | - Shir Lubaton-Barshishat
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Beer-Sheva, Israel
| | - Offer Erez
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Beer-Sheva, Israel
- Department of Obstetrics and Gynecology, Hutzel Women's Hospital, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
12
|
Lindley KJ, Perry A, Jacobs M, Petty L, Amancherla K, Zhao S, Barker C, Davila-Roman VG, Khan SS, Osmundson SS, Tanriverdi K, Freedman JE, Below J, Shah RV, Laurent LC. Differences in Cardiometabolic Proteins in Pregnancy Prioritize Relevant Targets of Preeclampsia. Arterioscler Thromb Vasc Biol 2024; 44:969-975. [PMID: 38385288 DOI: 10.1161/atvbaha.124.320737] [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: 01/17/2024] [Accepted: 02/05/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Preeclampsia is a hypertensive disorder of pregnancy characterized by widespread vascular inflammation. It occurs frequently in pregnancy, often without known risk factors, and has high rates of maternal and fetal morbidity and mortality. Identification of biomarkers that predict preeclampsia and its cardiovascular sequelae before clinical onset, or even before pregnancy, is a critical unmet need for the prevention of adverse pregnancy outcomes. METHODS We explored differences in cardiovascular proteomics (Olink Explore 384) in 256 diverse pregnant persons across 2 centers (26% Hispanic, 21% Black). RESULTS We identified significant differences in plasma abundance of markers associated with angiogenesis, blood pressure, cell adhesion, inflammation, and metabolism between individuals delivering with preeclampsia and controls, some of which have not been widely described previously and are not represented in the preeclampsia placental transcriptome. While we observed a broadly similar pattern in early (<34 weeks) versus late (≥34 weeks) preeclampsia, several proteins related to hemodynamic stress, hemostasis, and immune response appeared to be more highly dysregulated in early preeclampsia relative to late preeclampsia. CONCLUSIONS These results demonstrate the value of performing targeted proteomics using a panel of cardiovascular biomarkers to identify biomarkers relevant to preeclampsia pathophysiology and highlight the need for larger multiomic studies to define modifiable pathways of surveillance and intervention upstream to preeclampsia diagnosis.
Collapse
Affiliation(s)
- Kathryn J Lindley
- Vanderbilt Translational and Clinical Research Center, Cardiovascular Division (K.J.L., A.P., K.A., S.Z., K.T., J.E.F., R.V.S.), Vanderbilt University Medical Center, Nashville, TN
- Department of Obstetrics and Gynecology (K.J.L., S.S.O.), Vanderbilt University Medical Center, Nashville, TN
| | - Andrew Perry
- Vanderbilt Translational and Clinical Research Center, Cardiovascular Division (K.J.L., A.P., K.A., S.Z., K.T., J.E.F., R.V.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Marni Jacobs
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal Fetal Medicine, University of California San Diego (M.J.)
| | - Lauren Petty
- Division of Genetic Medicine (L.P., J.B.), Vanderbilt University Medical Center, Nashville, TN
| | - Kaushik Amancherla
- Vanderbilt Translational and Clinical Research Center, Cardiovascular Division (K.J.L., A.P., K.A., S.Z., K.T., J.E.F., R.V.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Shilin Zhao
- Vanderbilt Translational and Clinical Research Center, Cardiovascular Division (K.J.L., A.P., K.A., S.Z., K.T., J.E.F., R.V.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Claire Barker
- Cardiovascular Imaging and Clinical Research Core Laboratory, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (C.B., V.G.D.-R.)
| | - Victor G Davila-Roman
- Cardiovascular Imaging and Clinical Research Core Laboratory, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (C.B., V.G.D.-R.)
| | - Sadiya S Khan
- Cardiovascular Division, Feinberg School of Medicine, Northwestern University, Chicago, IL (S.S.K.)
| | - Sarah S Osmundson
- Department of Obstetrics and Gynecology (K.J.L., S.S.O.), Vanderbilt University Medical Center, Nashville, TN
| | - Kahraman Tanriverdi
- Vanderbilt Translational and Clinical Research Center, Cardiovascular Division (K.J.L., A.P., K.A., S.Z., K.T., J.E.F., R.V.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Jane E Freedman
- Vanderbilt Translational and Clinical Research Center, Cardiovascular Division (K.J.L., A.P., K.A., S.Z., K.T., J.E.F., R.V.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Jennifer Below
- Division of Genetic Medicine (L.P., J.B.), Vanderbilt University Medical Center, Nashville, TN
| | - Ravi V Shah
- Vanderbilt Translational and Clinical Research Center, Cardiovascular Division (K.J.L., A.P., K.A., S.Z., K.T., J.E.F., R.V.S.), Vanderbilt University Medical Center, Nashville, TN
| | | |
Collapse
|
13
|
Püschl IC, Thaneswaran Vyramuthu M, Bonde L, Lebech M, Iraqi Møller H, Vauvert F Hviid T, Lund Sørensen B, Macklon NS. Is salivary uric acid, a putative biomarker of pre-eclampsia, of maternal, placental, or fetal origin? Eur J Obstet Gynecol Reprod Biol 2024; 295:34-41. [PMID: 38330864 DOI: 10.1016/j.ejogrb.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 01/29/2024] [Accepted: 02/02/2024] [Indexed: 02/10/2024]
Abstract
OBJECTIVES Increased salivary uric acid (sUA) represents a potential biomarker predictive of pre-eclampsia (PE), but its origin is unclear. The study explores whether sUA levels reflect maternal or feto-placental physiological stress and whether sUA levels in these cases correlate with amniotic fluid (fetal origin), maternal blood (maternal origin), or cord blood (fetal vs placental origin). STUDY DESIGN Pregnant women (n = 39) undergoing amniotomy or caesarean section after 34 gestational weeks were designated into three groups of either maternal, feto-placental, or no signs of physiological stress: women (n = 15) in the established first phase of active labour and without any signs of fetal growth restriction (FGR) or PE were assigned to the maternal stress group, women (n = 6) with an ultrasound-based diagnosis of FGR, with or without PE, were assigned to the feto-placental stress group, and women (n = 18) not yet in active labour and without any signs of FGR or PE, were assigned to the control group. Uric acid levels in corresponding samples of amniotic fluid, saliva, maternal blood, and cord blood were compared between groups and between body compartments within each group. RESULTS The feto-placental stress group showed increased UA levels in saliva (median, interquartile range [IQR]: 0.47 [0.38] mmol/L, P = 0.023) and maternal blood (0.42 [0.13] mmol/L, P = 0.032), but no differences in amniotic fluid or cord blood compared with the other groups. Within the control and maternal stress group, sUA levels were lower compared with maternal blood (0.20 [0.08] vs 0.25 [0.08] mmol/L, Pcontrol = 0.018; 0.20 [0.06] vs 0.26 [0.08] mmol/L, Pmaternal = 0.001) and highest in amniotic fluid (control group (0.49 [0.18] mmol/L): Pmaternal,blood = 0.001, Pumbilical,artery = <0.001, Pumbilical,vein = <0.001, Psaliva = <0.001) (maternal stress group (0.56 [0.23] mmol/L): Pmaternal,blood = 0.021, Pumbilical,artery = 0.006, Pumbilical,vein = 0.004, Psaliva = 0.003). Levels did not differ between compartments in the feto-placental stress group. CONCLUSIONS Salivary and maternal blood UA levels were increased in the feto-placental stress group with salivary levels increasing more than blood levels compared with the maternal stress and control groups, whilst UA in amniotic fluid were not different between the groups, suggesting a placental origin and potential use of sUA as a biomarker of placental dysfunction, including FGR and severe PE.
Collapse
Affiliation(s)
- Ida Catharina Püschl
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; Department of Obstetrics and Gynaecology and ReproHealth Research Consortium, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark.
| | - Meera Thaneswaran Vyramuthu
- Department of Obstetrics and Gynaecology and ReproHealth Research Consortium, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark
| | - Lisbeth Bonde
- Department of Obstetrics and Gynaecology, Juliane Marie Centre, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Morten Lebech
- Department of Obstetrics and Gynaecology and ReproHealth Research Consortium, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark
| | - Hiba Iraqi Møller
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; Department of Clinical Biochemistry and ReproHealth Research Consortium, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark
| | - Thomas Vauvert F Hviid
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; Department of Clinical Biochemistry and ReproHealth Research Consortium, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark
| | - Bjarke Lund Sørensen
- Department of Obstetrics and Gynaecology and ReproHealth Research Consortium, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark
| | - Nicholas S Macklon
- Department of Obstetrics and Gynaecology and ReproHealth Research Consortium, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark; London Women's Clinic, 113-115 Harley St, London W1G 6AP, United Kingdom
| |
Collapse
|
14
|
Xia Y, Wang Y, Yuan S, Hu J, Zhang L, Xie J, Zhao Y, Hao J, Ren Y, Wu S. Development and validation of nomograms to predict clinical outcomes of preeclampsia. Front Endocrinol (Lausanne) 2024; 15:1292458. [PMID: 38549768 PMCID: PMC10972945 DOI: 10.3389/fendo.2024.1292458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/14/2024] [Indexed: 04/02/2024] Open
Abstract
Background Preeclampsia (PE) is one of the most severe pregnancy-related diseases; however, there is still a lack of reliable biomarkers. In this study, we aimed to develop models for predicting early-onset PE, severe PE, and the gestation duration of patients with PE. Methods Eligible patients with PE were enrolled and divided into a training (n = 253) and a validation (n = 108) cohort. Multivariate logistic and Cox models were used to identify factors associated with early-onset PE, severe PE, and the gestation duration of patients with PE. Based on significant factors, nomograms were developed and evaluated using the area under the curve (AUC) and a calibration curve. Results In the training cohort, multiple gravidity experience (p = 0.005), lower albumin (ALB; p < 0.001), and higher lactate dehydrogenase (LDH; p < 0.001) were significantly associated with early-onset PE. Abortion history (p = 0.017), prolonged thrombin time (TT; p < 0.001), and higher aspartate aminotransferase (p = 0.002) and LDH (p = 0.003) were significantly associated with severe PE. Abortion history (p < 0.001), gemellary pregnancy (p < 0.001), prolonged TT (p < 0.001), higher mean platelet volume (p = 0.014) and LDH (p < 0.001), and lower ALB (p < 0.001) were significantly associated with shorter gestation duration. Three nomograms were developed and validated to predict the probability of early-onset PE, severe PE, and delivery time for each patient with PE. The AUC showed good predictive performance, and the calibration curve and decision curve analysis demonstrated clinical practicability. Conclusion Based on the clinical features and peripheral blood laboratory indicators, we identified significant factors and developed models to predict early-onset PE, severe PE, and the gestation duration of pregnant women with PE, which could help clinicians assess the clinical outcomes early and design appropriate strategies for patients.
Collapse
Affiliation(s)
- Yan Xia
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Yao Wang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Shijin Yuan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaming Hu
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Lu Zhang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Jiamin Xie
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Yang Zhao
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Jiahui Hao
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yanwei Ren
- Department of Gynaecology and Obstetrics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shengjun Wu
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| |
Collapse
|
15
|
Gál L, Fóthi Á, Orosz G, Nagy S, Than NG, Orbán TI. Exosomal small RNA profiling in first-trimester maternal blood explores early molecular pathways of preterm preeclampsia. Front Immunol 2024; 15:1321191. [PMID: 38455065 PMCID: PMC10917917 DOI: 10.3389/fimmu.2024.1321191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/25/2024] [Indexed: 03/09/2024] Open
Abstract
Introduction Preeclampsia (PE) is a severe obstetrical syndrome characterized by new-onset hypertension and proteinuria and it is often associated with fetal intrauterine growth restriction (IUGR). PE leads to long-term health complications, so early diagnosis would be crucial for timely prevention. There are multiple etiologies and subtypes of PE, and this heterogeneity has hindered accurate identification in the presymptomatic phase. Recent investigations have pointed to the potential role of small regulatory RNAs in PE, and these species, which travel in extracellular vesicles (EVs) in the circulation, have raised the possibility of non-invasive diagnostics. The aim of this study was to investigate the behavior of exosomal regulatory small RNAs in the most severe subtype of PE with IUGR. Methods We isolated exosomal EVs from first-trimester peripheral blood plasma samples of women who later developed preterm PE with IUGR (n=6) and gestational age-matched healthy controls (n=14). The small RNA content of EVs and their differential expression were determined by next-generation sequencing and further validated by quantitative real-time PCR. We also applied the rigorous exceRpt bioinformatics pipeline for small RNA identification, followed by target verification and Gene Ontology analysis. Results Overall, >2700 small RNAs were identified in all samples and, of interest, the majority belonged to the RNA interference (RNAi) pathways. Among the RNAi species, 16 differentially expressed microRNAs were up-regulated in PE, whereas up-regulated and down-regulated members were equally found among the six identified Piwi-associated RNAs. Gene ontology analysis of the predicted small RNA targets showed enrichment of genes in pathways related to immune processes involved in decidualization, placentation and embryonic development, indicating that dysregulation of the induced small RNAs is connected to the impairment of immune pathways in preeclampsia development. Finally, the subsequent validation experiments revealed that the hsa_piR_016658 piRNA is a promising biomarker candidate for preterm PE associated with IUGR. Discussion Our rigorously designed study in a homogeneous group of patients unraveled small RNAs in circulating maternal exosomes that act on physiological pathways dysregulated in preterm PE with IUGR. Therefore, our small RNA hits are not only suitable biomarker candidates, but the revealed biological pathways may further inform us about the complex pathology of this severe PE subtype.
Collapse
Affiliation(s)
- Luca Gál
- Gene Regulation Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Ábel Fóthi
- Gene Regulation Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Gergő Orosz
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Sándor Nagy
- Department of Obstetrics and Gynecology, Petz Aladár University Teaching Hospital, Győr, Hungary
- Faculty of Health and Sport Sciences, Széchenyi István University, Győr, Hungary
| | - 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
- Genesis Theranostix Group, Budapest, Hungary
| | - Tamás I. Orbán
- Gene Regulation Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
16
|
Than NG, Romero R, Posta M, Györffy D, Szalai G, Rossi SW, Szilágyi A, Hupuczi P, Nagy S, Török O, Tarca AL, Erez O, Ács N, Papp Z. Classification of preeclampsia according to molecular clusters with the goal of achieving personalized prevention. J Reprod Immunol 2024; 161:104172. [PMID: 38141514 PMCID: PMC11027116 DOI: 10.1016/j.jri.2023.104172] [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: 07/10/2023] [Revised: 10/13/2023] [Accepted: 11/23/2023] [Indexed: 12/25/2023]
Abstract
The prevention of pre-eclampsia is difficult due to the syndromic nature and multiple underlying mechanisms of this severe complication of pregnancy. The current clinical distinction between early- and late-onset disease, although clinically useful, does not reflect the true nature and complexity of the pathologic processes leading to pre-eclampsia. The current gaps in knowledge on the heterogeneous molecular pathways of this syndrome and the lack of adequate, specific diagnostic methods are major obstacles to early screening and tailored preventive strategies. The development of novel diagnostic tools for detecting the activation of the identified disease pathways would enable early, accurate screening and personalized preventive therapies. We implemented a holistic approach that includes the utilization of different proteomic profiling methods of maternal plasma samples collected from various ethnic populations and the application of systems biology analysis to plasma proteomic, maternal demographic, clinical characteristic, and placental histopathologic data. This approach enabled the identification of four molecular subclasses of pre-eclampsia in which distinct and shared disease mechanisms are activated. The current review summarizes the results and conclusions from these studies and the research and clinical implications of our findings.
Collapse
Affiliation(s)
- Nándor Gábor Than
- Systems Biology of Reproduction Research Group, Institute of Enzymology, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary; Department of Obstetrics and Gynecology, School of Medicine, Semmelweis University, Budapest, Hungary; Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary; Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Genesis Theranostix Group, Budapest, Hungary.
| | - Roberto Romero
- Pregnancy Research Branch(1), NICHD/NIH/DHHS, 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
| | - Máté Posta
- Systems Biology of Reproduction Research Group, Institute of Enzymology, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary; Genesis Theranostix Group, Budapest, Hungary; Semmelweis University Doctoral School, Budapest, Hungary
| | - Dániel Györffy
- Systems Biology of Reproduction Research Group, Institute of Enzymology, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary; Genesis Theranostix Group, Budapest, Hungary; Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Gábor Szalai
- Systems Biology of Reproduction Research Group, Institute of Enzymology, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary; Genesis Theranostix Group, Budapest, Hungary; Department of Surgery, School of Medicine, University of Pécs, Pécs, Hungary
| | | | - András Szilágyi
- Systems Biology of Reproduction Research Group, Institute of Enzymology, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Petronella Hupuczi
- Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary; Department of Anesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
| | - Sándor Nagy
- Faculty of Health and Sport Sciences, Széchenyi István University, Győr, Hungary
| | - Olga Török
- Department of Obstetrics and Gynecology, School of Medicine, University of Debrecen, Debrecen, Hungary
| | - Adi L Tarca
- Genesis Theranostix Group, Budapest, Hungary; Pregnancy Research Branch(1), NICHD/NIH/DHHS, Bethesda, MD, USA; Department of Obstetrics and Gynecology, School of Medicine, Wayne State University School of Medicine, Detroit, MI, USA; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Offer Erez
- Genesis Theranostix Group, Budapest, Hungary; Pregnancy Research Branch(1), NICHD/NIH/DHHS, Bethesda, MD, USA; Department of Obstetrics and Gynecology, School of Medicine, Wayne State University School of Medicine, Detroit, MI, USA; Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben Gurion University of the Negev, Be'er Sheva, Israel
| | - Nándor Ács
- Department of Obstetrics and Gynecology, School of Medicine, Semmelweis University, Budapest, Hungary
| | - Zoltán Papp
- Department of Obstetrics and Gynecology, School of Medicine, Semmelweis University, Budapest, Hungary; Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary
| |
Collapse
|
17
|
Zhou S, Li J, Yang W, Xue P, Yin Y, Wang Y, Tian P, Peng H, Jiang H, Xu W, Huang S, Zhang R, Wei F, Sun HX, Zhang J, Zhao L. Noninvasive preeclampsia prediction using plasma cell-free RNA signatures. Am J Obstet Gynecol 2023; 229:553.e1-553.e16. [PMID: 37211139 DOI: 10.1016/j.ajog.2023.05.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/02/2023] [Accepted: 05/14/2023] [Indexed: 05/23/2023]
Abstract
BACKGROUND Preeclampsia, especially preterm preeclampsia and early-onset preeclampsia, is a life-threating pregnancy disorder, and the heterogeneity and complexity of preeclampsia make it difficult to predict risk and to develop treatments. Plasma cell-free RNA carries unique information from human tissue and may be useful for noninvasive monitoring of maternal, placental, and fetal dynamics during pregnancy. OBJECTIVE This study aimed to investigate various RNA biotypes associated with preeclampsia in plasma and to develop classifiers to predict preterm preeclampsia and early-onset preeclampsia before diagnosis. STUDY DESIGN We performed a novel, cell-free RNA sequencing method termed polyadenylation ligation-mediated sequencing to investigate the cell-free RNA characteristics of 715 healthy pregnancies and 202 pregnancies affected by preeclampsia before symptom onset. We explored differences in the abundance of different RNA biotypes in plasma between healthy and preeclampsia samples and built preterm preeclampsia and early-onset preeclampsia prediction classifiers using machine learning methods. Furthermore, we validated the performance of the classifiers using the external and internal validation cohorts and assessed the area under the curve and positive predictive value. RESULTS We detected 77 genes, including messenger RNA (44%) and microRNA (26%), that were differentially expressed in healthy mothers and mothers with preterm preeclampsia before symptom onset, which could separate participants with preterm preeclampsia from healthy samples and that played critical functional roles in preeclampsia physiology. We developed 2 classifiers for predicting preterm preeclampsia and early-onset preeclampsia before diagnosis based on 13 cell-free RNA signatures and 2 clinical features (in vitro fertilization and mean arterial pressure), respectively. Notably, both classifiers showed enhanced performance when compared with the existing methods. The preterm preeclampsia prediction model achieved 81% area under the curve and 68% positive predictive value in an independent validation cohort (preterm, n=46; control, n=151); the early-onset preeclampsia prediction model had an area under the curve of 88% and a positive predictive value of 73% in an external validation cohort (early-onset preeclampsia, n=28; control, n=234). Furthermore, we demonstrated that downregulation of microRNAs may play vital roles in preeclampsia through the upregulation of preeclampsia-relevant target genes. CONCLUSION In this cohort study, a comprehensive transcriptomic landscape of different RNA biotypes in preeclampsia was presented and 2 advanced classifiers with substantial clinical importance for preterm preeclampsia and early-onset preeclampsia prediction before symptom onset were developed. We demonstrated that messenger RNA, microRNA, and long noncoding RNA can simultaneously serve as potential biomarkers of preeclampsia, holding the promise of prevention of preeclampsia in the future. Abnormal cell-free messenger RNA, microRNA, and long noncoding RNA molecular changes may help to elucidate the pathogenic determinants of preeclampsia and open new therapeutic windows to effectively reduce pregnancy complications and fetal morbidity.
Collapse
Affiliation(s)
- Si Zhou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China; BGI Genomics, BGI-Shenzhen, Shenzhen, China; Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Shijiazhuang BGI Genomics Co, Ltd, Shijiazhuang, Hebei Province, China; Shijiazhuang BGI Clinical Laboratory Co, Ltd, Shijiazhuang, Hebei Province, China
| | - Jie Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China; BGI-Shenzhen, Shenzhen, China; BGI-Beijing, Beijing, China
| | - Wenzhi Yang
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Shijiazhuang BGI Genomics Co, Ltd, Shijiazhuang, Hebei Province, China; Shijiazhuang BGI Clinical Laboratory Co, Ltd, Shijiazhuang, Hebei Province, China
| | - Penghao Xue
- Shijiazhuang BGI Clinical Laboratory Co, Ltd, Shijiazhuang, Hebei Province, China
| | - Yanning Yin
- Shijiazhuang BGI Clinical Laboratory Co, Ltd, Shijiazhuang, Hebei Province, China
| | - Yunfang Wang
- Shijiazhuang BGI Clinical Laboratory Co, Ltd, Shijiazhuang, Hebei Province, China
| | | | | | | | - Wenqiu Xu
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Shang Huang
- Shenzhen Children's Hospital of China Medical University, Shenzhen, China
| | - Rui Zhang
- Division of Maternal-Fetal Medicine, Jinan University-affiliated Shenzhen Baoan Women's and Children's Hospital, Shenzhen, China.
| | - Fengxiang Wei
- Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Shenzhen, China.
| | - Hai-Xi Sun
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China; BGI-Shenzhen, Shenzhen, China; BGI-Beijing, Beijing, China.
| | - Jianguo Zhang
- BGI Genomics, BGI-Shenzhen, Shenzhen, China; Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Shijiazhuang BGI Genomics Co, Ltd, Shijiazhuang, Hebei Province, China; Shijiazhuang BGI Clinical Laboratory Co, Ltd, Shijiazhuang, Hebei Province, China.
| | - Lijian Zhao
- BGI Genomics, BGI-Shenzhen, Shenzhen, China; Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Shijiazhuang BGI Genomics Co, Ltd, Shijiazhuang, Hebei Province, China; Medical Technology College of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
18
|
Thomas G, Syngelaki A, Hamed K, Perez-Montaño A, Panigassi A, Tuytten R, Nicolaides KH. Preterm preeclampsia screening using biomarkers: combining phenotypic classifiers into robust prediction models. Am J Obstet Gynecol MFM 2023; 5:101110. [PMID: 37752025 DOI: 10.1016/j.ajogmf.2023.101110] [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: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND Preeclampsia screening is a critical component of antenatal care worldwide. Currently, the most developed screening test for preeclampsia at 11 to 13 weeks' gestation integrates maternal demographic characteristics and medical history with 3 biomarkers-serum placental growth factor, mean arterial pressure, and uterine artery pulsatility index-to identify approximately 75% of women who develop preterm preeclampsia with delivery before 37 weeks of gestation. It is generally accepted that further improvements to preeclampsia screening require the use of additional biomarkers. We recently reported that the levels of specific metabolites and metabolite ratios are associated with preterm preeclampsia. Notably, for several of these markers, preterm preeclampsia prediction varied according to maternal body mass index class. These findings motivated us to study whether patient classification allowed for combining metabolites with the current biomarkers more effectively to improve prediction of preterm preeclampsia. OBJECTIVE This study aimed to investigate whether metabolite biomarkers can improve biomarker-based preterm preeclampsia prediction in 3 screening resource scenarios according to the availability of: (1) placental growth factor, (2) placental growth factor+mean arterial pressure, and (3) placental growth factor+mean arterial pressure+uterine artery pulsatility index. STUDY DESIGN This was an observational case-control study, drawn from a large prospective screening study at 11 to 13 weeks' gestation on the prediction of pregnancy complications, conducted at King's College Hospital, London, United Kingdom. Maternal blood samples were also collected for subsequent research studies. We used liquid chromatography-mass spectrometry to quantify levels of 50 metabolites previously associated with pregnancy complications in plasma samples from singleton pregnancies. Biomarker data, normalized using multiples of medians, on 1635 control and 106 preterm preeclampsia pregnancies were available for model development. Modeling was performed using a methodology that generated a prediction model for preterm preeclampsia in 4 consecutive steps: (1) z-normalization of predictors, (2) combinatorial modeling of so-called (weak) classifiers in the unstratified patient set and in discrete patient strata based on body mass index and/or race, (3) selection of classifiers, and (4) aggregation of the selected classifiers (ie, bagging) into the final prediction model. The prediction performance of models was evaluated using the area under the receiver operating characteristic curve, and detection rate at 10% false-positive rate. RESULTS First, the predictor development methodology itself was evaluated. The patient set was split into a training set (2/3) and a test set (1/3) for predictor model development and internal validation. A prediction model was developed for each of the 3 different predictor panels, that is, placental growth factor+metabolites, placental growth factor+mean arterial pressure+metabolites, and placental growth factor+mean arterial pressure+uterine artery pulsatility index+metabolites. For all 3 models, the area under the receiver operating characteristic curve in the test set did not differ significantly from that of the training set. Next, a prediction model was developed using the complete data set for the 3 predictor panels. Among the 50 metabolites available for modeling, 26 were selected across the 3 prediction models; 21 contributed to at least 2 out of the 3 prediction models developed. Each time, area under the receiver operating characteristic curve and detection rate were significantly higher with the new prediction model than with the reference model. Markedly, the estimated detection rate with the placental growth factor+mean arterial pressure+metabolites prediction model in all patients was 0.58 (95% confidence interval, 0.49-0.70), a 15% increase (P<.001) over the detection rate of 0.43 (95% confidence interval, 0.33-0.55) estimated for the reference placental growth factor+mean arterial pressure. The same prediction model significantly improved detection in Black (14%) and White (19%) patients, and in the normal-weight group (18.5≤body mass index<25) and the obese group (body mass index≥30), with respectively 19% and 20% more cases detected, but not in the overweight group, when compared with the reference model. Similar improvement patterns in detection rates were found in the other 2 scenarios, but with smaller improvement amplitudes. CONCLUSION Metabolite biomarkers can be combined with the established biomarkers of placental growth factor, mean arterial pressure, and uterine artery pulsatility index to improve the biomarker component of early-pregnancy preterm preeclampsia prediction tests. Classification of the pregnant women according to the maternal characteristics of body mass index and/or race proved instrumental in achieving improved prediction. This suggests that maternal phenotyping can have a role in improving the prediction of obstetrical syndromes such as preeclampsia.
Collapse
Affiliation(s)
- Grégoire Thomas
- SQU4RE, Lokeren, Belgium (Dr Thomas); Metabolomic Diagnostics, Cork, Ireland (Drs Thomas, Panigassi, and Tuytten)
| | - Argyro Syngelaki
- The Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom (Drs Syngelaki, Hamed, Perez-Montaño, and Nicolaides)
| | - Karam Hamed
- The Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom (Drs Syngelaki, Hamed, Perez-Montaño, and Nicolaides)
| | - Anais Perez-Montaño
- The Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom (Drs Syngelaki, Hamed, Perez-Montaño, and Nicolaides)
| | - Ana Panigassi
- Metabolomic Diagnostics, Cork, Ireland (Drs Thomas, Panigassi, and Tuytten)
| | - Robin Tuytten
- Metabolomic Diagnostics, Cork, Ireland (Drs Thomas, Panigassi, and Tuytten).
| | - Kypros H Nicolaides
- The Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom (Drs Syngelaki, Hamed, Perez-Montaño, and Nicolaides)
| |
Collapse
|
19
|
Barak O, Lovelace T, Piekos S, Chu T, Cao Z, Sadovsky E, Mouillet JF, Ouyang Y, Parks WT, Hood L, Price ND, Benos PV, Sadovsky Y. Integrated unbiased multiomics defines disease-independent placental clusters in common obstetrical syndromes. BMC Med 2023; 21:349. [PMID: 37679695 PMCID: PMC10485945 DOI: 10.1186/s12916-023-03054-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Placental dysfunction, a root cause of common syndromes affecting human pregnancy, such as preeclampsia (PE), fetal growth restriction (FGR), and spontaneous preterm delivery (sPTD), remains poorly defined. These common, yet clinically disparate obstetrical syndromes share similar placental histopathologic patterns, while individuals within each syndrome present distinct molecular changes, challenging our understanding and hindering our ability to prevent and treat these syndromes. METHODS Using our extensive biobank, we identified women with severe PE (n = 75), FGR (n = 40), FGR with a hypertensive disorder (FGR + HDP; n = 33), sPTD (n = 72), and two uncomplicated control groups, term (n = 113), and preterm without PE, FGR, or sPTD (n = 16). We used placental biopsies for transcriptomics, proteomics, metabolomics data, and histological evaluation. After conventional pairwise comparison, we deployed an unbiased, AI-based similarity network fusion (SNF) to integrate the datatypes and identify omics-defined placental clusters. We used Bayesian model selection to compare the association between the histopathological features and disease conditions vs SNF clusters. RESULTS Pairwise, disease-based comparisons exhibited relatively few differences, likely reflecting the heterogeneity of the clinical syndromes. Therefore, we deployed the unbiased, omics-based SNF method. Our analysis resulted in four distinct clusters, which were mostly dominated by a specific syndrome. Notably, the cluster dominated by early-onset PE exhibited strong placental dysfunction patterns, with weaker injury patterns in the cluster dominated by sPTD. The SNF-defined clusters exhibited better correlation with the histopathology than the predefined disease groups. CONCLUSIONS Our results demonstrate that integrated omics-based SNF distinctively reclassifies placental dysfunction patterns underlying the common obstetrical syndromes, improves our understanding of the pathological processes, and could promote a search for more personalized interventions.
Collapse
Affiliation(s)
- Oren Barak
- Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 300 Halket Street, Pittsburgh, PA, 15213, USA
| | - Tyler Lovelace
- Department of Computational and Systems Biology, University of Pittsburgh, 800 Murdoch Building, 3420 Forbes Avenue, Pittsburgh, PA, 15260, USA
- Joint CMU-Pitt PhD Program in Computational Biology, 800 Murdoch Building, 3420 Forbes Avenue, Pittsburgh, PA, 15260, USA
| | - Samantha Piekos
- Institute for Systems Biology, 401 Terri Avenue North, Seattle, WA, 98109, USA
| | - Tianjiao Chu
- Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 300 Halket Street, Pittsburgh, PA, 15213, USA
| | - Zhishen Cao
- Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Elena Sadovsky
- Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Jean-Francois Mouillet
- Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 300 Halket Street, Pittsburgh, PA, 15213, USA
| | - Yingshi Ouyang
- Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 300 Halket Street, Pittsburgh, PA, 15213, USA
| | - W Tony Parks
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Simcoe Hall, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Leroy Hood
- Institute for Systems Biology, 401 Terri Avenue North, Seattle, WA, 98109, USA
| | - Nathan D Price
- Institute for Systems Biology, 401 Terri Avenue North, Seattle, WA, 98109, USA
- Thorne HealthTech, 152 West 57th Street, New York, NY, 10019, USA
| | - Panayiotis V Benos
- Department of Computational and Systems Biology, University of Pittsburgh, 800 Murdoch Building, 3420 Forbes Avenue, Pittsburgh, PA, 15260, USA
- Joint CMU-Pitt PhD Program in Computational Biology, 800 Murdoch Building, 3420 Forbes Avenue, Pittsburgh, PA, 15260, USA
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, University of Florida, 2004 Mowry Road, Gainesville, FL, 32610, USA
| | - Yoel Sadovsky
- Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213, USA.
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 300 Halket Street, Pittsburgh, PA, 15213, USA.
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
20
|
Xie Y, Zhao F, Freitag N, Borowski S, Wang Y, Harms C, Pang PC, Desforges J, Wen T, Schwedhelm E, Singh M, Dechend R, Dell A, Haslam SM, Dveksler G, Garcia MG, Blois SM. Maternal-derived galectin-1 shapes the placenta niche through Sda terminal glycosylation: Implication for preeclampsia. PNAS NEXUS 2023; 2:pgad247. [PMID: 37575671 PMCID: PMC10416815 DOI: 10.1093/pnasnexus/pgad247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/20/2023] [Indexed: 08/15/2023]
Abstract
Placental abnormalities cause impaired fetal growth and poor pregnancy outcome (e.g. preeclampsia [PE]) with long-lasting consequences for the mother and offspring. The molecular dialogue between the maternal niche and the developing placenta is critical for the function of this organ. Galectin-1 (gal-1), a highly expressed glycan-binding protein at the maternal-fetal interface, orchestrates the maternal adaptation to pregnancy and placenta development. Down-regulation or deficiency of gal-1 during pregnancy is associated with the development of PE; however, the maternal- and placental-derived gal-1 contributions to the disease onset are largely unknown. We demonstrate that lack of gal-1 imposes a risk for PE development in a niche-specific manner, and this is accompanied by a placental dysfunction highly influenced by the absence of maternal-derived gal-1. Notably, differential placental glycosylation through the Sda-capped N-glycans dominates the invasive trophoblast capacity triggered by maternal-derived gal-1. Our findings show that gal-1 derived from the maternal niche is essential for healthy placenta development and indicate that impairment of the gal-1 signaling pathway within the maternal niche could be a molecular cause for maternal cardiovascular maladaptation during pregnancy.
Collapse
Affiliation(s)
- Yiran Xie
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Fangqi Zhao
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Nancy Freitag
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max-Delbrück Center for Molecular Medicine (MDC) and Charité-Universitätsmedizin, 13125 Berlin, Germany
| | - Sophia Borowski
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max-Delbrück Center for Molecular Medicine (MDC) and Charité-Universitätsmedizin, 13125 Berlin, Germany
| | - Yiru Wang
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Charlotte Harms
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Poh-Choo Pang
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Juliette Desforges
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Tianyu Wen
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf and German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20249 Hamburg, Germany
| | - Manvendra Singh
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, 37075 Göttingen, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max-Delbrück Center for Molecular Medicine (MDC) and Charité-Universitätsmedizin, 13125 Berlin, Germany
- Department of Cardiology and Nephrology, HELIOS-Klinikum, 13125 Berlin, Germany
| | - Anne Dell
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Stuart M Haslam
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Gabriela Dveksler
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Mariana G Garcia
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Sandra M Blois
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
21
|
Bai Y, Li S. Oxidative Stress Sensing System for 8-OHdG Detection Based on Plasma Coupled Electrochemistry by Transparent ITO/AuNTAs/PtNPs Electrode. BIOSENSORS 2023; 13:643. [PMID: 37367008 PMCID: PMC10296443 DOI: 10.3390/bios13060643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/01/2023] [Accepted: 06/09/2023] [Indexed: 06/28/2023]
Abstract
8-Hydroxydeoxyguanosine (8-OHdG) is the most widely used oxidative stress biomarker of the free radical-induced oxidative damage product of DNA, which may allow a premature assessment of various diseases. This paper designs a label-free, portable biosensor device to directly detect 8-OHdG by plasma-coupled electrochemistry on a transparent and conductive indium tin oxide (ITO) electrode. We reported a flexible printed ITO electrode made from particle-free silver and carbon inks. After inkjet printing, the working electrode was sequentially assembled by gold nanotriangles (AuNTAs) and platinum nanoparticles (PtNPs). This nanomaterial-modified portable biosensor showed excellent electrochemical performance for 8-OHdG detection from 10 μg/mL to 100 μg/mL by our self-developed constant voltage source integrated circuit system. This work demonstrated a portable biosensor for simultaneously integrating nanostructure, electroconductivity, and biocompatibility to construct advanced biosensors for oxidative damage biomarkers. The proposed nanomaterial-modified ITO-based electrochemical portable device was a potential biosensor to approach 8-OHdG point-of-care testing (POCT) in various biological fluid samples, such as saliva and urine samples.
Collapse
Affiliation(s)
| | - Shuang Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China;
| |
Collapse
|
22
|
Erez O, Gotsch F, Jung E, Chaiworapongsa T, Gudicha DW, Suksai M, Gallo DM, Chaemsaithong P, Bosco M, Al Qasem M, Meyyazhagan A, Than NG, Romero R. Perturbations in kinetics of the thrombin generation assay identify women at risk of preeclampsia in the first trimester and provide the rationale for a preventive approach. Am J Obstet Gynecol 2023; 228:580.e1-580.e17. [PMID: 36368431 PMCID: PMC10149548 DOI: 10.1016/j.ajog.2022.11.1276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Activation of the coagulation system and increased thrombin generation have been implicated in the pathophysiology of preeclampsia, and this rationale supports the administration of low-molecular-weight heparin to prevent this syndrome in patients at risk. Yet, randomized trials of this prophylactic measure have yielded contradictory results. A possible explanation is that only a subset of patients with preeclampsia have excessive thrombin generation and would benefit from the administration of low-molecular-weight heparin. Therefore, the key questions are whether and when patients who subsequently develop preeclampsia present evidence of abnormal thrombin generation. OBJECTIVE This study aimed to determine (1) the kinetics of thrombin generation throughout gestation in women with a normal pregnancy and in those with early and late preeclampsia, and (2) the diagnostic performance of in vivo thrombin generation parameters to predict the development of preeclampsia. STUDY DESIGN This retrospective, nested case-control study was based on a prospective longitudinal cohort of singleton gestations. Cases comprised women who developed preeclampsia (n=49), and controls consisted of patients with a normal pregnancy (n=45). Preeclampsia was classified into early-onset (n=24) and late-onset (n=25). Longitudinal changes in the parameters of the thrombin generation assay (lag time, time to peak thrombin concentration, peak thrombin concentration, endogenous thrombin generation, and velocity index) throughout gestation were compared between the study groups, and normal pregnancy percentiles were derived from the control group. We tested whether a single parameter or a combination of parameters, derived from the kinetics of thrombin generation, could identify patients who subsequently developed preeclampsia. Time-related parameters <10th percentile were considered short, and concentration-related parameters >90th percentile were considered high. RESULTS (1) Patients who developed preeclampsia (early- and late-onset) had abnormal thrombin generation kinetics as early as 8 to 16 weeks of pregnancy; (2) patients with a combination of a short lag time and high peak thrombin concentration at 8 to 16 weeks of pregnancy had an odds ratio of 43.87 for the subsequent development of preeclampsia (area under the curve, 0.79; sensitivity, 56.8%; specificity, 92.7%; positive likelihood ratio, 7.76); (3) at 16 to 22 weeks of gestation, patients with a combination of a short lag time and a high velocity index had an odds ratio of 16 for the subsequent development of preeclampsia (area under the curve, 0.78; sensitivity, 62.2%; specificity, 92.5%; positive likelihood ratio, 8.29). CONCLUSION During early pregnancy, the thrombin generation assay can identify the subset of patients at a greater risk for the development of preeclampsia owing to accelerated and enhanced production of thrombin. This observation provides a rationale for testing the efficacy of low-molecular-weight heparin in this subset of patients. We propose that future research on the efficacy of low-molecular-weight heparin and other interventions targeting the coagulation system to prevent preeclampsia should be focused on patients with abnormal kinetics of thrombin generation.
Collapse
Affiliation(s)
- Offer Erez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Obstetrics and Gynecology, Emek Medical Center, Afula, Israel
| | - 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, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - 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, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, 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, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Dereje W Gudicha
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - 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, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - 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, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Piya Chaemsaithong
- Perinatology Research Branch, 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, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Mariachiara Bosco
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Malek Al Qasem
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Arun Meyyazhagan
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Nandor Gabor Than
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary; Maternity Private Clinic, Budapest, Hungary; First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI; Detroit Medical Center, Detroit, MI.
| |
Collapse
|
23
|
Han L, Holland OJ, Da Silva Costa F, Perkins AV. Potential biomarkers for late-onset and term preeclampsia: A scoping review. Front Physiol 2023; 14:1143543. [PMID: 36969613 PMCID: PMC10036383 DOI: 10.3389/fphys.2023.1143543] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
Preeclampsia is a progressive, multisystem pregnancy disorder. According to the time of onset or delivery, preeclampsia has been subclassified into early-onset (<34 weeks) and late-onset (≥34 weeks), or preterm (<37 weeks) and term (≥37 weeks). Preterm preeclampsia can be effectively predicted at 11-13 weeks well before onset, and its incidence can be reduced by preventively using low-dose aspirin. However, late-onset and term preeclampsia are more prevalent than early forms and still lack effective predictive and preventive measures. This scoping review aims to systematically identify the evidence of predictive biomarkers reported in late-onset and term preeclampsia. This study was conducted based on the guidance of the Joanna Briggs Institute (JBI) methodology for scoping reviews. The Preferred Reporting Items for Systematic Reviews and Meta-Analysis extension for scoping reviews (PRISMA-ScR) was used to guide the study. The following databases were searched for related studies: PubMed, Web of Science, Scopus, and ProQuest. Search terms contain "preeclampsia," "late-onset," "term," "biomarker," or "marker," and other synonyms combined as appropriate using the Boolean operators "AND" and "OR." The search was restricted to articles published in English from 2012 to August 2022. Publications were selected if study participants were pregnant women and biomarkers were detected in maternal blood or urine samples before late-onset or term preeclampsia diagnosis. The search retrieved 4,257 records, of which 125 studies were included in the final assessment. The results demonstrate that no single molecular biomarker presents sufficient clinical sensitivity and specificity for screening late-onset and term preeclampsia. Multivariable models combining maternal risk factors with biochemical and/or biophysical markers generate higher detection rates, but they need more effective biomarkers and validation data for clinical utility. This review proposes that further research into novel biomarkers for late-onset and term preeclampsia is warranted and important to find strategies to predict this complication. Other critical factors to help identify candidate markers should be considered, such as a consensus on defining preeclampsia subtypes, optimal testing time, and sample types.
Collapse
Affiliation(s)
- Luhao Han
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Olivia J. Holland
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Fabricio Da Silva Costa
- Maternal Fetal Medicine Unit, Gold Coast University Hospital, Gold Coast, QLD, Australia
- School of Medicine and Dentistry, Griffith University, Gold Coast, QLD, Australia
| | - Anthony V. Perkins
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
- School of Health, University of the Sunshine Coast, Sunshine Coast, QLD, Australia
| |
Collapse
|
24
|
Abstract
Pre-eclampsia is a life-threatening disease of pregnancy unique to humans and a leading cause of maternal and neonatal morbidity and mortality. Women who survive pre-eclampsia have reduced life expectancy, with increased risks of stroke, cardiovascular disease and diabetes, while babies from a pre-eclamptic pregnancy have increased risks of preterm birth, perinatal death and neurodevelopmental disability and cardiovascular and metabolic disease later in life. Pre-eclampsia is a complex multisystem disease, diagnosed by sudden-onset hypertension (>20 weeks of gestation) and at least one other associated complication, including proteinuria, maternal organ dysfunction or uteroplacental dysfunction. Pre-eclampsia is found only when a placenta is or was recently present and is classified as preterm (delivery <37 weeks of gestation), term (delivery ≥37 weeks of gestation) and postpartum pre-eclampsia. The maternal syndrome of pre-eclampsia is driven by a dysfunctional placenta, which releases factors into maternal blood causing systemic inflammation and widespread maternal endothelial dysfunction. Available treatments target maternal hypertension and seizures, but the only 'cure' for pre-eclampsia is delivery of the dysfunctional placenta and baby, often prematurely. Despite decades of research, the aetiology of pre-eclampsia, particularly of term and postpartum pre-eclampsia, remains poorly defined. Significant advances have been made in the prediction and prevention of preterm pre-eclampsia, which is predicted in early pregnancy through combined screening and is prevented with daily low-dose aspirin, starting before 16 weeks of gestation. By contrast, the prediction of term and postpartum pre-eclampsia is limited and there are no preventive treatments. Future research must investigate the pathogenesis of pre-eclampsia, in particular of term and postpartum pre-eclampsia, and evaluate new prognostic tests and treatments in adequately powered clinical trials.
Collapse
|
25
|
Than NG, Romero R, Györffy D, Posta M, Bhatti G, Done B, Chaemsaithong P, Jung E, Suksai M, Gotsch F, Gallo DM, Bosco M, Kim B, Kim YM, Chaiworapongsa T, Rossi SW, Szilágyi A, Erez O, Tarca AL, Papp Z. Molecular subclasses of preeclampsia characterized by a longitudinal maternal proteomics study: distinct biomarkers, disease pathways and options for prevention. J Perinat Med 2023; 51:51-68. [PMID: 36253935 PMCID: PMC9837387 DOI: 10.1515/jpm-2022-0433] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 09/09/2022] [Indexed: 01/25/2023]
Abstract
OBJECTIVES The heterogeneous nature of preeclampsia is a major obstacle to early screening and prevention, and a molecular taxonomy of disease is needed. We have previously identified four subclasses of preeclampsia based on first-trimester plasma proteomic profiles. Herein, we expanded this approach by using a more comprehensive panel of proteins profiled in longitudinal samples. METHODS Proteomic data collected longitudinally from plasma samples of women who developed preeclampsia (n=109) and of controls (n=90) were available from our previous report on 1,125 proteins. Consensus clustering was performed to identify subgroups of patients with preeclampsia based on data from five gestational-age intervals by using select interval-specific features. Demographic, clinical, and proteomic differences among clusters were determined. Differentially abundant proteins were used to identify cluster-specific perturbed KEGG pathways. RESULTS Four molecular clusters with different clinical phenotypes were discovered by longitudinal proteomic profiling. Cluster 1 involves metabolic and prothrombotic changes with high rates of early-onset preeclampsia and small-for-gestational-age neonates; Cluster 2 includes maternal anti-fetal rejection mechanisms and recurrent preeclampsia cases; Cluster 3 is associated with extracellular matrix regulation and comprises cases of mostly mild, late-onset preeclampsia; and Cluster 4 is characterized by angiogenic imbalance and a high prevalence of early-onset disease. CONCLUSIONS This study is an independent validation and further refining of molecular subclasses of preeclampsia identified by a different proteomic platform and study population. The results lay the groundwork for novel diagnostic and personalized tools of prevention.
Collapse
Affiliation(s)
- Nándor Gábor Than
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
- Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary
- Genesis Theranostix Group, Budapest, Hungary
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, Michigan, USA
| | - Dániel Györffy
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
- Genesis Theranostix Group, Budapest, Hungary
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Máté Posta
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
- Genesis Theranostix Group, Budapest, Hungary
- Károly Rácz Doctoral School of Clinical Medicine, Semmelweis University, Budapest, Hungary
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Bogdan Done
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Piya Chaemsaithong
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - 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, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - 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, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - 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, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - 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, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Universidad Del Valle, Cali, Colombia
| | - Mariachiara Bosco
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Bomi Kim
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Yeon Mee Kim
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - 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, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | - András Szilágyi
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Offer Erez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Genesis Theranostix Group, Budapest, Hungary
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, HaEmek Medical Center, Afula, Israel
| | - Adi L. Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Genesis Theranostix Group, Budapest, Hungary
- 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
| | - Zoltán Papp
- Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
26
|
Oravecz O, Romero R, Tóth E, Kapitány J, Posta M, Gallo DM, Rossi SW, Tarca AL, Erez O, Papp Z, Matkó J, Than NG, Balogh A. Placental galectins regulate innate and adaptive immune responses in pregnancy. Front Immunol 2022; 13:1088024. [PMID: 36643922 PMCID: PMC9832025 DOI: 10.3389/fimmu.2022.1088024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/05/2022] [Indexed: 12/29/2022] Open
Abstract
Introduction Galectins are master regulators of maternal immune responses and placentation in pregnancy. Galectin-13 (gal-13) and galectin-14 (gal-14) are expressed solely by the placenta and contribute to maternal-fetal immune tolerance by inducing the apoptosis of activated T lymphocytes and the polarization of neutrophils toward an immune-regulatory phenotype.Furthermore, their decreased placental expression is associated with pregnancy complications, such as preeclampsia and miscarriage. Yet, our knowledge of the immunoregulatory role of placental galectins is incomplete. Methods This study aimed to investigate the effects of recombinant gal-13 and gal-14 on cell viability, apoptosis, and cytokine production of peripheral blood mononuclear cells (PBMCs) and the signaling pathways involved. Results Herein, we show that gal-13 and gal-14 bind to the surface of non-activated PBMCs (monocytes, natural killer cells, B cells, and T cells) and increase their viability while decreasing the rate of their apoptosis without promoting cell proliferation. We also demonstrate that gal-13 and gal-14 induce the production of interleukin (IL)-8, IL-10, and interferon-gamma cytokines in a concentration-dependent manner in PBMCs. The parallel activation of Erk1/2, p38, and NF-ĸB signaling evidenced by kinase phosphorylation in PBMCs suggests the involvement of these pathways in the regulation of the galectin-affected immune cell functions. Discussion These findings provide further evidence on how placenta-specific galectins assist in the establishment and maintenance of a proper immune environment during a healthy pregnancy.
Collapse
Affiliation(s)
- Orsolya Oravecz
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary,Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, United States,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, United States,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States,Detroit Medical Center, Detroit, MI, United States
| | - Eszter Tóth
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Judit Kapitány
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Máté Posta
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary,Károly Rácz Doctoral School of Clinical Medicine, Semmelweis University, Budapest, Hungary
| | - Dahiana M. Gallo
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States,Department of Obstetrics and Gynecology, Universidad Del Valle, Cali, Colombia
| | | | - Adi L. Tarca
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States,Genesis Theranostix Group, Budapest, Hungary
| | - Offer Erez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States,Genesis Theranostix Group, Budapest, Hungary,Department of Obstetrics and Gynecology, Soroka University Medical Center, Beer Sheva, Israel
| | - Zoltán Papp
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary,Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary
| | - János Matkó
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Nándor Gábor Than
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary,Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States,Genesis Theranostix Group, Budapest, Hungary,Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary,Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary,*Correspondence: Nándor Gábor Than,
| | - Andrea Balogh
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| |
Collapse
|
27
|
Oravecz O, Balogh A, Romero R, Xu Y, Juhasz K, Gelencser Z, Xu Z, Bhatti G, Pique-Regi R, Peterfia B, Hupuczi P, Kovalszky I, Murthi P, Tarca AL, Papp Z, Matko J, Than NG. Proteoglycans: Systems-Level Insight into Their Expression in Healthy and Diseased Placentas. Int J Mol Sci 2022; 23:5798. [PMID: 35628608 PMCID: PMC9147780 DOI: 10.3390/ijms23105798] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 02/04/2023] Open
Abstract
Proteoglycan macromolecules play key roles in several physiological processes (e.g., adhesion, proliferation, migration, invasion, angiogenesis, and apoptosis), all of which are important for placentation and healthy pregnancy. However, their precise roles in human reproduction have not been clarified. To fill this gap, herein, we provide an overview of the proteoglycans' expression and role in the placenta, in trophoblast development, and in pregnancy complications (pre-eclampsia, fetal growth restriction), highlighting one of the most important members of this family, syndecan-1 (SDC1). Microarray data analysis showed that of 34 placentally expressed proteoglycans, SDC1 production is markedly the highest in the placenta and that SDC1 is the most upregulated gene during trophoblast differentiation into the syncytiotrophoblast. Furthermore, placental transcriptomic data identified dysregulated proteoglycan genes in pre-eclampsia and in fetal growth restriction, including SDC1, which is supported by the lower concentration of syndecan-1 in maternal blood in these syndromes. Overall, our clinical and in vitro studies, data analyses, and literature search pointed out that proteoglycans, as important components of the placenta, may regulate various stages of placental development and participate in the maintenance of a healthy pregnancy. Moreover, syndecan-1 may serve as a useful marker of syncytialization and a prognostic marker of adverse pregnancy outcomes. Further studies are warranted to explore the role of proteoglycans in healthy and complicated pregnancies, which may help in diagnostic or therapeutic developments.
Collapse
Affiliation(s)
- Orsolya Oravecz
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
- Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Andrea Balogh
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Detroit Medical Center, Detroit, MI 48201, USA
| | - Yi Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Kata Juhasz
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
| | - Zsolt Gelencser
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
| | - Zhonghui Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Roger Pique-Regi
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Balint Peterfia
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
| | | | - Ilona Kovalszky
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary;
| | - Padma Murthi
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia;
- Department of Obstetrics and Gynaecology, University of Melbourne, Royal Women’s Hospital, Parkville, VIC 3502, Australia
| | - Adi L. Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI 48202, USA
| | - Zoltan Papp
- Maternity Private Clinic, H-1126 Budapest, Hungary; (P.H.); (Z.P.)
| | - Janos Matko
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
| | - Nandor Gabor Than
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
- Maternity Private Clinic, H-1126 Budapest, Hungary; (P.H.); (Z.P.)
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary;
| |
Collapse
|