1
|
Jahan F, Vasam G, Cariaco Y, Nik-Akhtar A, Green A, Menzies KJ, Bainbridge SA. NAD + depletion is central to placental dysfunction in an inflammatory subclass of preeclampsia. Life Sci Alliance 2024; 7:e202302505. [PMID: 39389781 PMCID: PMC11467044 DOI: 10.26508/lsa.202302505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/12/2024] Open
Abstract
Preeclampsia (PE) is a hypertensive disorder of pregnancy and a major cause of maternal/perinatal adverse health outcomes with no effective therapeutic strategies. Our group previously identified distinct subclasses of PE, one of which exhibits heightened placental inflammation (inflammation-driven PE). In non-pregnant populations, chronic inflammation is associated with decreased levels of cellular NAD+, a vitamin B3 derivative involved in energy metabolism and mitochondrial function. Interestingly, specifically in placentas from women with inflammation-driven PE, we observed the increased activity of NAD+-consuming enzymes, decreased NAD+ content, decreased expression of mitochondrial proteins, and increased oxidative damage. HTR8 human trophoblasts likewise demonstrated increased NAD+-dependent ADP-ribosyltransferase (ART) activity, coupled with decreased mitochondrial respiration rates and invasive function under inflammatory conditions. Such adverse effects were attenuated by boosting cellular NAD+ levels with nicotinamide riboside (NR). Finally, in an LPS-induced rat model of inflammation-driven PE, NR administration (200 mg/kg/day) from gestational days 1-19 prevented maternal hypertension and fetal/placental growth restriction, improved placental mitochondrial function, and reduced inflammation and oxidative stress. This study demonstrates the critical role of NAD+ in maintaining placental function and identifies NAD+ boosting as a promising preventative strategy for PE.
Collapse
Affiliation(s)
- Fahmida Jahan
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Goutham Vasam
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Canada
| | - Yusmaris Cariaco
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Canada
| | - Abolfazl Nik-Akhtar
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Alex Green
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Keir J Menzies
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Canada
| | - Shannon A Bainbridge
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
2
|
Yuen N, Lemaire M, Wilson SL. Cell-free placental DNA: What do we really know? PLoS Genet 2024; 20:e1011484. [PMID: 39652523 PMCID: PMC11627368 DOI: 10.1371/journal.pgen.1011484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Abstract
Cell-free placental DNA (cfpDNA) is present in maternal circulation during gestation. CfpDNA carries great potential as a research and clinical tool as it provides a means to investigate the placental (epi)genome across gestation, which previously required invasive placenta sampling procedures. CfpDNA has been widely implemented in the clinical setting for noninvasive prenatal testing (NIPT). Despite this, the basic biology of cfpDNA remains poorly understood, limiting the research and clinical utility of cfpDNA. This review will examine the current knowledge of cfpDNA, including origins and molecular characteristics, highlight gaps in knowledge, and discuss future research directions.
Collapse
Affiliation(s)
- Natalie Yuen
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Melanie Lemaire
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Samantha L. Wilson
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
3
|
Trummer O, Stern C, Reintar S, Mayer-Pickel K, Cervar-Zivkovic M, Dischinger U, Kurlbaum M, Huppertz B, Fluhr H, Obermayer-Pietsch B. Steroid Profiles and Precursor-to-Product Ratios Are Altered in Pregnant Women with Preeclampsia. Int J Mol Sci 2024; 25:12704. [PMID: 39684415 DOI: 10.3390/ijms252312704] [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: 10/30/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Steroid hormone imbalance is associated with the pathogenesis of preeclampsia. However, affected enzymes of steroid metabolism and gene and protein expression in serum and placenta have not been elucidated yet. We aimed to investigate steroid hormone profiles and precursor-to-product ratios in preeclamptic women compared to women with healthy pregnancy (controls) to identify potentially affected steroid hormones and their metabolizing enzymes. Also, we aimed to investigate whether the mRNA expression of these enzymes is different between the study groups and whether levels of serum mRNA expression reflect postnatal placental protein expression. Serum levels of 14 steroid hormones were measured at eight time points throughout pregnancy in nine preeclamptic women and 36 controls. Serum mRNA expression of selected steroid-metabolizing enzymes was assessed, and their protein expression was analyzed in additional nine preeclamptic women. Mean levels of sex steroid and corticosteroid hormones were significantly altered in preeclamptic women. Precursor-to-product ratios of 5α-reductase, aromatase and 11β-hydroxysteroid dehydrogenase 1 were significantly increased, those of steroid 17α-hydroxylase, 17β-hydroxysteroid-dehydrogenase, steroid 11β-hydroxylase and 11β-hydroxysteroid dehydrogenase 2 were significantly decreased. Serum mRNA expression and placenta protein expression were comparable between the groups. Results contribute to understanding the heterogeneity of preeclampsia and can thus promote future research in personalized medicine.
Collapse
Affiliation(s)
- Olivia Trummer
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria
| | - Christina Stern
- Department of Obstetrics and Gynecology, Division of Obstetrics, Medical University of Graz, 8036 Graz, Austria
| | - Sharmaine Reintar
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria
| | - Karoline Mayer-Pickel
- Department of Obstetrics and Gynecology, Division of Obstetrics, Medical University of Graz, 8036 Graz, Austria
| | - Mila Cervar-Zivkovic
- Department of Obstetrics and Gynecology, Division of Obstetrics, Medical University of Graz, 8036 Graz, Austria
| | - Ulrich Dischinger
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, 97080 Würzburg, Germany
- Core Unit Clinical Mass Spectrometry, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Max Kurlbaum
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, 97080 Würzburg, Germany
- Core Unit Clinical Mass Spectrometry, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Berthold Huppertz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Herbert Fluhr
- Department of Obstetrics and Gynecology, Division of Obstetrics, Medical University of Graz, 8036 Graz, Austria
| | - Barbara Obermayer-Pietsch
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria
| |
Collapse
|
4
|
Jacobs A, Al-Juboori SI, Dobrinskikh E, Bolt MA, Sammel MD, Lijewski V, Post MD, Small JM, Su EJ. Placental differences between severe fetal growth restriction and hypertensive disorders of pregnancy requiring early preterm delivery: morphometric analysis of the villous tree supported by artificial intelligence. Am J Obstet Gynecol 2024; 231:552.e1-552.e13. [PMID: 38423447 PMCID: PMC11347726 DOI: 10.1016/j.ajog.2024.02.291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND The great obstetrical syndromes of fetal growth restriction and hypertensive disorders of pregnancy can occur individually or be interrelated. Placental pathologic findings often overlap between these conditions, regardless of whether 1 or both diagnoses are present. Quantification of placental villous structures in each of these settings may identify distinct differences in developmental pathways. OBJECTIVE This study aimed to determine how the quantity and surface area of placental villi and vessels differ between severe, early-onset fetal growth restriction with absent or reversed umbilical artery Doppler indices and hypertensive disorders of pregnancy or the 2 conditions combined among subjects with disease severity that warrant early preterm delivery. We hypothesized that the trajectories of placental morphogenesis diverge after a common initiating insult of deep defective placentation. Specifically, we postulated that only villi are affected in pregnancy-related hypertension, whereas both villous and vascular structures are proportionally diminished in severe fetal growth restriction with no additional effect when hypertension is concomitantly present. STUDY DESIGN In this retrospective cohort study, paraffin-embedded placental tissue was obtained from 4 groups, namely (1) patients with severe fetal growth restriction with absent or reversed umbilical artery end-diastolic velocities and hypertensive disorders of pregnancy, (2) patients with severe fetal growth restriction with absent or reversed umbilical artery Doppler indices and no hypertension, (3) gestational age-matched, appropriately grown pregnancies with hypertensive disease, and (4) gestational age-matched, appropriately grown pregnancies without hypertension. Dual immunohistochemistry for cytokeratin-7 (trophoblast) and CD34 (endothelial cells) was performed, followed by artificial intelligence-driven morphometric analyses. The number of villi, total villous area, number of fetoplacental vessels, and total vascular area across villi within a uniform region of interest were quantified. Quantitative analyses of placental structures were modeled using linear regression. RESULTS Placentas from pregnancies complicated by hypertensive disorders of pregnancy exhibited significantly fewer stem villi (-282 stem villi; 95% confidence interval, -467 to -98; P<.01), a smaller stem villous area (-4.3 mm2; 95% confidence interval, -7.3 to -1.2; P<.01), and fewer stem villous vessels (-4967 stem villous vessels; 95% confidence interval, -8501 to -1433; P<.01) with no difference in the total vascular area. In contrast, placental abnormalities in cases with severe growth restriction were limited to terminal villi with global decreases in the number of villi (-873 terminal villi; 95% confidence interval, -1501 to -246; P<.01), the villous area (-1.5 mm2; 95% confidence interval, -2.7 to -0.4; P<.01), the number of blood vessels (-5165 terminal villous vessels; 95% confidence interval, -8201 to -2128; P<.01), and the vascular area (-0.6 mm2; 95% confidence interval, -1.1 to -0.1; P=.02). The combination of hypertension and growth restriction had no additional effect beyond the individual impact of each state. CONCLUSION Pregnancies complicated by hypertensive disorders of pregnancy exhibited defects in the stem villi only, whereas placental abnormalities in severely growth restricted pregnancies with absent or reversed umbilical artery end-diastolic velocities were limited to the terminal villi. There were no significant statistical interactions in the combination of growth restriction and hypertension, suggesting that distinct pathophysiological pathways downstream of the initial insult of defective placentation are involved in each entity and do not synergize to lead to more severe pathologic consequences. Delineating mechanisms that underly the divergence in placental development after a common inciting event of defective deep placentation may shed light on new targets for prevention or treatment.
Collapse
Affiliation(s)
- Anna Jacobs
- Rocky Vista University College of Osteopathic Medicine, Parker, CO
| | - Saif I Al-Juboori
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Evgenia Dobrinskikh
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO
| | - Matthew A Bolt
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO
| | - Mary D Sammel
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO
| | - Virginia Lijewski
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO
| | - Miriam D Post
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO
| | - James M Small
- Department of Biomedical Sciences; Rocky Vista University College of Osteopathic Medicine, Parker, CO
| | - Emily J Su
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO; Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO.
| |
Collapse
|
5
|
Massri N, Arora R. Uterine stromal but not epithelial PTGS2 is critical for murine pregnancy success. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.620133. [PMID: 39484555 PMCID: PMC11527190 DOI: 10.1101/2024.10.24.620133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Use of non-steroidal anti-inflammatory drugs that target prostaglandin synthase (PTGS) enzymes have been implicated in miscarriage. Further, PTGS2-derived prostaglandins are reduced in the endometrium of patients with a history of implantation failure. However, in the mouse model of pregnancy, peri-implantation PTGS2 function is controversial. Some studies suggest that Ptgs2-/- mice display deficits in ovulation, fertilization, and implantation, while other studies suggest a role for PTGS2 only in ovulation but not implantation. Further, the uterine cell type responsible for PTGS2 function and role of PTGS2 in regulating implantation chamber formation is not known. To address this we generated tissue-specific deletion models of Ptgs2. We observed that PTGS2 ablation from the epithelium alone in Ltfcre/+; Ptgs2f/f mice and in both the epithelium and endothelium of the Pax2cre/+; Ptgs2f/f mice does not affect embryo implantation. Further, deletion of PTGS2 in the ovary, oviduct, and the uterus using Pgrcre/+; Ptgs2f/f does not disrupt pre-implantation events but instead interferes with post-implantation chamber formation, vascular remodeling and decidualization. While all embryos initiate chamber formation, more than half of the embryos fail to transition from blastocyst to epiblast stage, resulting in embryo death and resorbing decidual sites at mid-gestation. Thus, our results suggest no role for uterine epithelial PTGS2 in early pregnancy but instead highlight a role for uterine stromal PTGS2 in modulating post-implantation embryo and implantation chamber growth. Overall, our study provides clarity on the compartment-specific role of PTGS2 and provides a valuable model for further investigating the role of stromal PTGS2 in post-implantation embryo development.
Collapse
Affiliation(s)
- Noura Massri
- Cell and Molecular Biology Program, Michigan State University
- Institute for Quantitative Health Science and Engineering, Michigan State University
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University
| | - Ripla Arora
- Cell and Molecular Biology Program, Michigan State University
- Institute for Quantitative Health Science and Engineering, Michigan State University
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University
| |
Collapse
|
6
|
Couture C, Caron M, St-Onge P, Brien ME, Sinnett D, Dal Soglio D, Girard S. Identification of divergent placental profiles in clinically distinct pregnancy complications revealed by the transcriptome. Placenta 2024; 154:184-192. [PMID: 39042974 DOI: 10.1016/j.placenta.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Pregnancy complications, including preeclampsia (PE), preterm birth (PTB), and intra-uterine growth restriction (IUGR) have individually been associated with inflammation but the combined comparative analysis of their placental profiles at the transcriptomic and histological levels is lacking. METHODS Bulk RNA-sequencing of human placental biopsies from uncomplicated term pregnancies (CTL) and pregnancies complicated with early-onset (EO), and late-onset (LO) PE, as well as PTB and term IUGR were used to characterize individual molecular profiles. We also applied immune-cell-specific cellular deconvolution to address local immune cell compositions and analyzed placental lesions by histology to further characterize these complications. RESULTS Transcriptome analysis revealed that clinically distinct complications differentiated themselves in unique ways compared to CTLs. Only TMEM136 was commonly modulated. Compared to CTLs, we found that PTB and IUGR were the most distinct, with LOPE being the least distinct. PTB and IUGR revealed differently enhanced inflammatory pathways, where PTB had general inflammatory responses and IUGR had immune cell activation. This inflammation was reflected in the histological profile for PTB only, whereas structural lesions were elevated in all complications. Placental lesions additionally had corresponding enhancement in inflammatory and structural biological processes. We observed that having co-complications, particularly for PTB with or without IUGR, impacted placental transcriptomes. Lastly, cellular deconvolution uncovered shared immune features among the complications. DISCUSSION Overall, we provide evidence that these pregnancy complications are not only distinct in their clinical manifestations but also in their placental profiles, which could be leveraged to understand their underlying mechanisms and could offer therapeutic targets.
Collapse
Affiliation(s)
- Camille Couture
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada; Sainte-Justine Hospital Research Center, Montreal, QC, Canada
| | - Maxime Caron
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Université de Montréal, Montreal, Quebec, Canada
| | - Pascal St-Onge
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada
| | - Marie-Eve Brien
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada
| | - Daniel Sinnett
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada; Department of Pediatrics, Université de Montreal, Montreal, Quebec, Canada
| | - Dorothée Dal Soglio
- Department of Pathology and Cellular Biology, Université de Montréal, Montreal, QC, Canada
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Université de Montréal, Montreal, Quebec, Canada; Department of Obstetrics and Gynecology, Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
7
|
Bowman-Gibson S, Chandiramani C, Stone ML, Waker CA, Rackett TM, Maxwell RA, Dhanraj DN, Brown TL. Streamlined Analysis of Maternal Plasma Indicates Small Extracellular Vesicles are Significantly Elevated in Early-Onset Preeclampsia. Reprod Sci 2024; 31:2771-2782. [PMID: 38777947 PMCID: PMC11393201 DOI: 10.1007/s43032-024-01591-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
Preeclampsia (PE) is a leading cause of maternal and fetal mortality and morbidity. While placental dysfunction is a core underlying issue, the pathogenesis of this disorder is thought to differ between early-onset (EOPE) and late-onset (LOPE) subtypes. As recent reports suggest that small extracellular vesicles (sEVs) contribute to the development of PE, we have compared systemic sEV concentrations between normotensive, EOPE, and LOPE pregnancies. To circumvent lengthy isolation techniques and intermediate filtration steps, a streamlined approach was developed to evaluate circulating plasma sEVs from maternal plasma. Polymer-based precipitation and purification were used to isolate total systemic circulating maternal sEVs, free from bias toward specific surface marker expression or extensive subpurification. Immediate Nanoparticle Tracking Analysis (NTA) of freshly isolated sEV samples afforded a comprehensive analysis that can be completed within hours, avoiding confounding freeze-thaw effects of particle aggregation and degradation.Rather than exosomal subpopulations, our findings indicate a significant elevation in the total number of circulating maternal sEVs in patients with EOPE. This streamlined approach also preserves sEV-bound protein and microRNA (miRNA) that can be used for potential biomarker analysis. This study is one of the first to demonstrate that maternal plasma sEVs harbor full-length hypoxia inducible factor 1 alpha (HIF-1α) protein, with EOPE sEVs carrying higher levels of HIF-1α compared to control sEVs. The detection of HIF-1α and its direct signaling partner microRNA-210 (miR-210) within systemic maternal sEVs lays the groundwork for identifying how sEV signaling contributes to the development of preeclampsia. When taken together, our quantitative and qualitative results provide compelling evidence to support the translational potential of streamlined sEV analysis for future use in the clinical management of patients with EOPE.
Collapse
Affiliation(s)
- Scout Bowman-Gibson
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, 3640 Colonel Glenn Highway, 457 NEC Building, Dayton, OH, 45435, USA
| | - Chandni Chandiramani
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, 3640 Colonel Glenn Highway, 457 NEC Building, Dayton, OH, 45435, USA
- Department of Obstetrics and Gynecology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45435, USA
| | - Madison L Stone
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, 3640 Colonel Glenn Highway, 457 NEC Building, Dayton, OH, 45435, USA
| | - Christopher A Waker
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, 3640 Colonel Glenn Highway, 457 NEC Building, Dayton, OH, 45435, USA
| | - Traci M Rackett
- Department of Obstetrics and Gynecology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45435, USA
| | - Rose A Maxwell
- Department of Obstetrics and Gynecology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45435, USA
| | - David N Dhanraj
- Department of Obstetrics and Gynecology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45435, USA
| | - Thomas L Brown
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, 3640 Colonel Glenn Highway, 457 NEC Building, Dayton, OH, 45435, USA.
- Department of Obstetrics and Gynecology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45435, USA.
| |
Collapse
|
8
|
Cameron K, Borahay M, Hong X, Baker V, Vaught A, Wang X. Uterine fibroids and risk of hypertensive disorders of pregnancy - results from a racially diverse high-risk cohort. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.05.24303830. [PMID: 38496516 PMCID: PMC10942496 DOI: 10.1101/2024.03.05.24303830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Study Question What is the impact of the presence of uterine fibroids on the risk of developing hypertensive disorders of pregnancy (HDP) in a predominantly urban, low-income, Black, and Hispanic population of women with ultrasound or clinically diagnosed uterine fibroids with rich phenotypic data to carefully control for potential confounders? Summary answers The odds of HDP were 39% higher in women with uterine fibroids compared to those without when controlled for age at delivery, race, prepregnancy BMI, education, parity, and smoking status; neither fibroid location or size modified this risk. What is known already Studies are conflicting regarding the impact of uterine fibroids on risk of HDP; limitations of prior studies include primarily Western European populations and lack of measurement of potential confounders. Study design size and duration A total of 7030 women from the Boston Birth Cohort (a racially diverse cohort recruited from 1998 to 2018) that had clinical and ultrasound data regarding uterine fibroid status were included in this analysis. Participants/materials setting and methods Four hundred eighty-nine women with uterine fibroids and 6541 women without were included. Hypertensive disorders of pregnancy were ascertained from medical records. Logistic regression was performed to assess the risk of HDP in women with and without uterine fibroids. Covariates adjusted for included age at delivery, race, pre-pregnancy BMI, education, parity, and smoking status during pregnancy. Sub-analyses were performed to assess the impact of specific fibroid location and overall fibroid volume burden. Main results and the role of chance The incidence of uterine fibroids in the cohort was 7% (N=489). Twelve percent of women without uterine fibroids and 17% of women with fibroids developed HDP; in multivariate analyses adjusted for the potential confounders above, the odds of HDP were 39% higher in women with uterine fibroids compared to those without (p=0.03). Women with a uterine fibroid diagnosis based on ICD code (n=297) versus asymptomatic incidental ultrasound diagnosis (n=192) had a significantly greater chance of developing HDP (20 vs 15%, p=0.006). There did not appear to be an association between number of fibroids or total fibroid volume and the risk of developing HDP. Limitations, reasons for caution: This study has a relatively small sample size. While post-hoc power calculation determined that there was adequate power to detect a 4.6% difference in the incidence of development of HDP between participants with uterine fibroids and those without, the sub-analyses based on fibroid size, location, and method of diagnosis were underpowered to determine a similar level of difference. Wider implications of the findings In a racially diverse cohort, presence of uterine fibroids was a significant risk factor for developing HDP, regardless of uterine fibroid size or location. This may have implications for additional monitoring and risk stratification in women with uterine fibroids. Study funding/competing interests KC supported by WRHR NIH NICHD Award # K12 HD103036, PI Andrew Satin, RD James Segars. The Boston Birth Cohort (the parent study) was supported in part by the National Institutes of Health (NIH) grants (2R01HD041702, R01HD098232, R01ES031272, R01ES031521, and U01 ES034983); and the Health Resources and Services Administration (HRSA) of the U.S. Department of Health and Human Services (HHS) (UT7MC45949). This information or content and conclusions are those of the authors and should not be construed as the official position or policy of, nor should any endorsements be inferred by any funding agencies. Trial registration number The BBC is registered under clinicaltrials.gov NCT03228875 .
Collapse
|
9
|
Youssef L, Testa L, Crovetto F, Crispi F. 10. Role of high dimensional technology in preeclampsia (omics in preeclampsia). Best Pract Res Clin Obstet Gynaecol 2024; 92:102427. [PMID: 37995432 DOI: 10.1016/j.bpobgyn.2023.102427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/05/2023] [Accepted: 08/06/2023] [Indexed: 11/25/2023]
Abstract
Preeclampsia is a pregnancy-specific disease that has no known precise cause. Integrative biology approach based on multi-omics has been applied to identify upstream pathways and better understand the pathophysiology of preeclampsia. At DNA level, genomics and epigenomics studies have revealed numerous genetic variants associated with preeclampsia, including those involved in regulating blood pressure and immune response. Transcriptomics analyses have revealed altered expression of genes in preeclampsia, particularly those related to inflammation and angiogenesis. At protein level, proteomics studies have identified potential biomarkers for preeclampsia diagnosis and prediction in addition to revealing the main pathophysiological pathways involved in this disease. At metabolite level, metabolomics has highlighted altered lipid and amino acid metabolisms in preeclampsia. Finally, microbiomics studies have identified dysbiosis in the gut and vaginal microbiota in pregnant women with preeclampsia. Overall, omics technologies have improved our understanding of the complex molecular mechanisms underlying preeclampsia. However, further research is warranted to fully integrate and translate these omics findings into clinical practice.
Collapse
Affiliation(s)
- Lina Youssef
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, IDIBAPS, University of Barcelona, Barcelona, Spain; Institut de Recerca August Pi Sunyer (IDIBAPS), Barcelona, Spain; Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain.
| | - Lea Testa
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Francesca Crovetto
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, IDIBAPS, University of Barcelona, Barcelona, Spain; Institut de Recerca Sant Joan de Deu (IRSJD), Barcelona, Spain
| | - Fatima Crispi
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, IDIBAPS, University of Barcelona, Barcelona, Spain; Institut de Recerca August Pi Sunyer (IDIBAPS), Barcelona, Spain; Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| |
Collapse
|
10
|
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
|
11
|
Kovacheva VP, Eberhard BW, Cohen RY, Maher M, Saxena R, Gray KJ. Preeclampsia Prediction Using Machine Learning and Polygenic Risk Scores From Clinical and Genetic Risk Factors in Early and Late Pregnancies. Hypertension 2024; 81:264-272. [PMID: 37901968 PMCID: PMC10842389 DOI: 10.1161/hypertensionaha.123.21053] [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: 02/07/2023] [Accepted: 10/12/2023] [Indexed: 10/31/2023]
Abstract
BACKGROUND Preeclampsia, a pregnancy-specific condition associated with new-onset hypertension after 20-weeks gestation, is a leading cause of maternal and neonatal morbidity and mortality. Predictive tools to understand which individuals are most at risk are needed. METHODS We identified a cohort of N=1125 pregnant individuals who delivered between May 2015 and May 2022 at Mass General Brigham Hospitals with available electronic health record data and linked genetic data. Using clinical electronic health record data and systolic blood pressure polygenic risk scores derived from a large genome-wide association study, we developed machine learning (XGBoost) and logistic regression models to predict preeclampsia risk. RESULTS Pregnant individuals with a systolic blood pressure polygenic risk score in the top quartile had higher blood pressures throughout pregnancy compared with patients within the lowest quartile systolic blood pressure polygenic risk score. In the first trimester, the most predictive model was XGBoost, with an area under the curve of 0.74. In late pregnancy, with data obtained up to the delivery admission, the best-performing model was XGBoost using clinical variables, which achieved an area under the curve of 0.91. Adding the systolic blood pressure polygenic risk score to the models did not improve the performance significantly based on De Long test comparing the area under the curve of models with and without the polygenic score. CONCLUSIONS Integrating clinical factors into predictive models can inform personalized preeclampsia risk and achieve higher predictive power than the current practice. In the future, personalized tools can be implemented to identify high-risk patients for preventative therapies and timely intervention to improve adverse maternal and neonatal outcomes.
Collapse
Affiliation(s)
- Vesela P Kovacheva
- Department of Anesthesiology, Perioperative and Pain Medicine (V.P.K., B.W.E., R.Y.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Braden W Eberhard
- Department of Anesthesiology, Perioperative and Pain Medicine (V.P.K., B.W.E., R.Y.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Raphael Y Cohen
- Department of Anesthesiology, Perioperative and Pain Medicine (V.P.K., B.W.E., R.Y.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- PathAI, Boston, MA (R.Y.C.)
| | - Matthew Maher
- Department of Anesthesia, Critical Care and Pain Medicine, Center for Genomic Medicine, Massachusetts General Hospital, Boston (M.M., R.S., K.J.G.)
| | - Richa Saxena
- Department of Anesthesia, Critical Care and Pain Medicine, Center for Genomic Medicine, Massachusetts General Hospital, Boston (M.M., R.S., K.J.G.)
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston (R.S.)
| | - Kathryn J Gray
- Division of Maternal-Fetal Medicine (K.J.G.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Department of Anesthesia, Critical Care and Pain Medicine, Center for Genomic Medicine, Massachusetts General Hospital, Boston (M.M., R.S., K.J.G.)
| |
Collapse
|
12
|
Giannubilo SR, Cecati M, Marzioni D, Ciavattini A. Circulating miRNAs and Preeclampsia: From Implantation to Epigenetics. Int J Mol Sci 2024; 25:1418. [PMID: 38338700 PMCID: PMC10855731 DOI: 10.3390/ijms25031418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
In this review, we comprehensively present the literature on circulating microRNAs (miRNAs) associated with preeclampsia, a pregnancy-specific disease considered the primary reason for maternal and fetal mortality and morbidity. miRNAs are single-stranded non-coding RNAs, 20-24 nt long, which control mRNA expression. Changes in miRNA expression can induce a variation in the relative mRNA level and influence cellular homeostasis, and the strong presence of miRNAs in all body fluids has made them useful biomarkers of several diseases. Preeclampsia is a multifactorial disease, but the etiopathogenesis remains unclear. The functions of trophoblasts, including differentiation, proliferation, migration, invasion and apoptosis, are essential for a successful pregnancy. During the early stages of placental development, trophoblasts are strictly regulated by several molecular pathways; however, an imbalance in these molecular pathways can lead to severe placental lesions and pregnancy complications. We then discuss the role of miRNAs in trophoblast invasion and in the pathogenesis, diagnosis and prediction of preeclampsia. We also discuss the potential role of miRNAs from an epigenetic perspective with possible future therapeutic implications.
Collapse
Affiliation(s)
| | - Monia Cecati
- Department of Clinical Sciences, Università Politecnica delle Marche, 60020 Ancona, Italy; (S.R.G.); (A.C.)
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy;
| | - Andrea Ciavattini
- Department of Clinical Sciences, Università Politecnica delle Marche, 60020 Ancona, Italy; (S.R.G.); (A.C.)
| |
Collapse
|
13
|
Patnaik P, Khodaee A, Vasam G, Mukherjee A, Salsabili S, Ukwatta E, Grynspan D, Chan ADC, Bainbridge S. Automated detection of microscopic placental features indicative of maternal vascular malperfusion using machine learning. Placenta 2024; 145:19-26. [PMID: 38011757 DOI: 10.1016/j.placenta.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023]
Abstract
INTRODUCTION Hypertensive disorders of pregnancy (HDP) and fetal growth restriction (FGR) are common obstetrical complications, often with pathological features of maternal vascular malperfusion (MVM) in the placenta. Currently, clinical placental pathology methods involve a manual visual examination of histology sections, a practice that can be resource-intensive and demonstrates moderate-to-poor inter-pathologist agreement on diagnostic outcomes, dependant on the degree of pathologist sub-specialty training. METHODS This study aims to apply machine learning (ML) feature extraction methods to classify digital images of placental histopathology specimens, collected from cases of HDP [pregnancy induced hypertension (PIH), preeclampsia (PE), PE + FGR], normotensive FGR, and healthy pregnancies, according to the presence or absence of MVM lesions. 159 digital images were captured from histological placental specimens, manually scored for MVM lesions (MVM- or MVM+) and used to develop a support vector machine (SVM) classifier model, using features extracted from pre-trained ResNet18. The model was trained with data augmentation and shuffling, with the performance assessed for patch-level and image-level classification through measurements of accuracy, precision, and recall using confusion matrices. RESULTS The SVM model demonstrated accuracies of 70 % and 79 % for patch-level and image-level MVM classification, respectively, with poorest performance observed on images with borderline MVM presence, as determined through post hoc observation. DISCUSSION The results are promising for the integration of ML methods into the placental histopathological examination process. Using this study as a proof-of-concept will lead our group and others to carry ML models further in placental histopathology.
Collapse
Affiliation(s)
- Purvasha Patnaik
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Afsoon Khodaee
- Department of Systems and Computer Engineering, Carleton University, Ottawa, ON, Canada
| | - Goutham Vasam
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Anika Mukherjee
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Sina Salsabili
- Department of Systems and Computer Engineering, Carleton University, Ottawa, ON, Canada
| | - Eranga Ukwatta
- Department of Systems and Computer Engineering, Carleton University, Ottawa, ON, Canada; School of Engineering, University of Guelph, Guelph, ON, Canada
| | - David Grynspan
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Children's Hospital of Eastern Ontario, Ottawa, ON, Canada; Department of Pathology and Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vernon Jubilee Hospital, Vancouver, BC, Canada
| | - Adrian D C Chan
- Department of Systems and Computer Engineering, Carleton University, Ottawa, ON, Canada
| | - Shannon Bainbridge
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
14
|
Khan SS, Petito LC, Huang X, Harrington K, McNeil RB, Bello NA, Merz CNB, Miller EC, Ravi R, Scifres C, Catov J, Pemberton V, Varagic J, Zee PC, Yee LM, Ray M, Kim JK, Lane-Cordova A, Lewey J, Theilen LH, Saade GR, Greenland P, Grobman WA. Body Mass Index, Adverse Pregnancy Outcomes, and Cardiovascular Disease Risk. Circ Res 2023; 133:725-735. [PMID: 37814889 PMCID: PMC10578703 DOI: 10.1161/circresaha.123.322762] [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: 03/07/2023] [Accepted: 09/08/2023] [Indexed: 10/11/2023]
Abstract
BACKGROUND Obesity is a well-established risk factor for both adverse pregnancy outcomes (APOs) and cardiovascular disease (CVD). However, it is not known whether APOs are mediators or markers of the obesity-CVD relationship. This study examined the association between body mass index, APOs, and postpartum CVD risk factors. METHODS The sample included adults from the nuMoM2b (Nulliparous Pregnancy Outcomes Study: Monitoring Mothers-To-Be) Heart Health Study who were enrolled in their first trimester (6 weeks-13 weeks 6 days gestation) from 8 United States sites. Participants had a follow-up visit at 3.7 years postpartum. APOs, which included hypertensive disorders of pregnancy, preterm birth, small-for-gestational-age birth, and gestational diabetes, were centrally adjudicated. Mediation analyses estimated the association between early pregnancy body mass index and postpartum CVD risk factors (hypertension, hyperlipidemia, and diabetes) and the proportion mediated by each APO adjusted for demographics and baseline health behaviors, psychosocial stressors, and CVD risk factor levels. RESULTS Among 4216 participants enrolled, mean±SD maternal age was 27±6 years. Early pregnancy prevalence of overweight was 25%, and obesity was 22%. Hypertensive disorders of pregnancy occurred in 15%, preterm birth in 8%, small-for-gestational-age birth in 11%, and gestational diabetes in 4%. Early pregnancy obesity, compared with normal body mass index, was associated with significantly higher incidence of postpartum hypertension (adjusted odds ratio, 1.14 [95% CI, 1.10-1.18]), hyperlipidemia (1.11 [95% CI, 1.08-1.14]), and diabetes (1.03 [95% CI, 1.01-1.04]) even after adjustment for baseline CVD risk factor levels. APOs were associated with higher incidence of postpartum hypertension (1.97 [95% CI, 1.61-2.40]) and hyperlipidemia (1.31 [95% CI, 1.03-1.67]). Hypertensive disorders of pregnancy mediated a small proportion of the association between obesity and incident hypertension (13% [11%-15%]) and did not mediate associations with incident hyperlipidemia or diabetes. There was no significant mediation by preterm birth or small-for-gestational-age birth. CONCLUSIONS There was heterogeneity across APO subtypes in their association with postpartum CVD risk factors and mediation of the association between early pregnancy obesity and postpartum CVD risk factors. However, only a small or nonsignificant proportion of the association between obesity and CVD risk factors was mediated by any of the APOs, suggesting APOs are a marker of prepregnancy CVD risk and not a predominant cause of postpartum CVD risk.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Rupa Ravi
- Columbia University Irving Medical Center
| | | | | | | | | | | | - Lynn M Yee
- Northwestern University Feinberg School of Medicine
| | - Mitali Ray
- University of Pittsburgh School of Medicine
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Jahan F, Vasam G, Cariaco Y, Nik-Akhtar A, Green A, Menzies KJ, Bainbridge SA. A comparison of rat models that best mimic immune-driven preeclampsia in humans. Front Endocrinol (Lausanne) 2023; 14:1219205. [PMID: 37842294 PMCID: PMC10569118 DOI: 10.3389/fendo.2023.1219205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/04/2023] [Indexed: 10/17/2023] Open
Abstract
Preeclampsia (PE), a hypertensive pregnancy disorder, can originate from varied etiology. Placenta malperfusion has long been considered the primary cause of PE. However, we and others have showed that this disorder can also result from heightened inflammation at the maternal-fetal interface. To advance our understanding of this understudied PE subtype, it is important to establish validated rodent models to study the pathophysiology and test therapies. We evaluated three previously described approaches to induce inflammation-mediated PE-like features in pregnant rats: 1) Tumor necrosis factor-α (TNF-α) infusion via osmotic pump from gestational day (GD) 14-19 at 50ng/day/animal; 2) Polyinosinic:polycytidylic acid (Poly I:C) intraperitoneal (IP) injections from GD 10-18 (alternate days) at 10mg/kg/day/animal; and, 3) Lipopolysaccharide (LPS) IP injections from GD 13-18 at 20ug-70ug/kg/day per animal. Maternal blood pressure was measured by tail-cuff. Upon sacrifice, fetal and placenta weights were recorded. Placenta histomorphology was assessed using H&E sections. Placenta inflammation was determined by quantifying TNF-α levels and inflammatory gene expression. Placenta metabolic and mitochondrial health were determined by measuring mitochondrial respiration rates and placenta NAD+/NADH content. Of the three rodent models tested, we found that Poly I:C and LPS decreased both fetal weight and survival; and correlated with a reduction in region specific placenta growth. As the least effective model characterized, TNF-α treatment resulted in a subtle decrease in fetal/placenta weight and placenta mitochondrial respiration. Only the LPS model was able to induce maternal hypertension and exhibited pronounced placenta metabolic and mitochondrial dysfunction, common features of PE. Thus, the rat LPS model was most effective for recapitulating features observed in cases of human inflammatory PE. Future mechanistic and/or therapeutic intervention studies focuses on this distinct PE patient population may benefit from the employment of this rodent model of PE.
Collapse
Affiliation(s)
- Fahmida Jahan
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Goutham Vasam
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Yusmaris Cariaco
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Abolfazl Nik-Akhtar
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Alex Green
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Keir J. Menzies
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Shannon A. Bainbridge
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
16
|
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
|
17
|
Fernández-Boyano I, Inkster AM, Yuan V, Robinson WP. eoPred: predicting the placental phenotype of early-onset preeclampsia using public DNA methylation data. Front Genet 2023; 14:1248088. [PMID: 37736302 PMCID: PMC10509376 DOI: 10.3389/fgene.2023.1248088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/08/2023] [Indexed: 09/23/2023] Open
Abstract
Background: A growing body of literature has reported molecular and histological changes in the human placenta in association with preeclampsia (PE). Placental DNA methylation (DNAme) and transcriptomic patterns have revealed molecular subgroups of PE that are associated with placental histopathology and clinical phenotypes of the disease. However, the clinical and molecular heterogeneity of PE both across and within subtypes complicates the study of this disease. PE is most strongly associated with placental pathology and adverse fetal and maternal outcomes when it develops early in pregnancy. We focused on placentae from pregnancies affected by preeclampsia that were delivered before 34 weeks of gestation to develop eoPred, a predictor of the DNAme signature associated with the placental phenotype of early-onset preeclampsia (EOPE). Results: Public data from 83 placental samples (HM450K), consisting of 42 EOPE and 41 normotensive preterm birth (nPTB) cases, was used to develop eoPred-a supervised model that relies on a highly discriminative 45 CpG DNAme signature of EOPE in the placenta. The performance of eoPred was assessed using cross-validation (AUC = 0.95) and tested in an independent validation cohort (n = 49, AUC = 0.725). A subset of fetal growth restriction (FGR) and late-PE cases showed a similar DNAme profile at the 45 predictive CpGs, consistent with the overlap in placental pathology between these conditions. The relationship between the EOPE probability generated by eoPred and various phenotypic variables was also assessed, revealing that it is associated with gestational age, and it is not driven by cell composition differences. Conclusion: eoPred relies on a 45-CpG DNAme signature to predict a homogeneous placental phenotype of EOPE in a discrete or continuous manner. Using this classifier should 1) aid in the study of placental insufficiency and improve the consistency of future placental DNAme studies of PE, 2) facilitate identifying the placental phenotype of EOPE in public data sets and 3) importantly, standardize the placental diagnosis of EOPE to allow better cross-cohort comparisons. Lastly, classification of cases with eoPred will be useful for investigating the relationship between placental pathology and genetic or environmental variables.
Collapse
Affiliation(s)
- I. Fernández-Boyano
- BC Children’s Hospital Research Institute (BCCHR), Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - A. M. Inkster
- BC Children’s Hospital Research Institute (BCCHR), Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - V. Yuan
- BC Children’s Hospital Research Institute (BCCHR), Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - W. P. Robinson
- BC Children’s Hospital Research Institute (BCCHR), Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
18
|
Chaemsaithong P, Gil MM, Chaiyasit N, Cuenca-Gomez D, Plasencia W, Rolle V, Poon LC. Accuracy of placental growth factor alone or in combination with soluble fms-like tyrosine kinase-1 or maternal factors in detecting preeclampsia in asymptomatic women in the second and third trimesters: a systematic review and meta-analysis. Am J Obstet Gynecol 2023; 229:222-247. [PMID: 36990308 DOI: 10.1016/j.ajog.2023.03.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/09/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023]
Abstract
OBJECTIVE This study aimed to: (1) identify all relevant studies reporting on the diagnostic accuracy of maternal circulating placental growth factor) alone or as a ratio with soluble fms-like tyrosine kinase-1), and of placental growth factor-based models (placental growth factor combined with maternal factors±other biomarkers) in the second or third trimester to predict subsequent development of preeclampsia in asymptomatic women; (2) estimate a hierarchical summary receiver-operating characteristic curve for studies reporting on the same test but different thresholds, gestational ages, and populations; and (3) select the best method to screen for preeclampsia in asymptomatic women during the second and third trimester of pregnancy by comparing the diagnostic accuracy of each method. DATA SOURCES A systematic search was performed through MEDLINE, Embase, CENTRAL, ClinicalTrials.gov, and the World Health Organization International Clinical Trials Registry Platform databases from January 1, 1985 to April 15, 2021. STUDY ELIGIBILITY CRITERIA Studies including asymptomatic singleton pregnant women at >18 weeks' gestation with risk of developing preeclampsia were evaluated. We included only cohort or cross-sectional test accuracy studies reporting on preeclampsia outcome, allowing tabulation of 2×2 tables, with follow-up available for >85%, and evaluating performance of placental growth factor alone, soluble fms-like tyrosine kinase-1- placental growth factor ratio, or placental growth factor-based models. The study protocol was registered on the International Prospective Register Of Systematic Reviews (CRD 42020162460). METHODS Because of considerable intra- and interstudy heterogeneity, we computed the hierarchical summary receiver-operating characteristic plots and derived diagnostic odds ratios, β, θi, and Λ for each method to compare performances. The quality of the included studies was evaluated by the QUADAS-2 tool. RESULTS The search identified 2028 citations, from which we selected 474 studies for detailed assessment of the full texts. Finally, 100 published studies met the eligibility criteria for qualitative and 32 for quantitative syntheses. Twenty-three studies reported on performance of placental growth factor testing for the prediction of preeclampsia in the second trimester, including 16 (with 27 entries) that reported on placental growth factor test alone, 9 (with 19 entries) that reported on the soluble fms-like tyrosine kinase-1-placental growth factor ratio, and 6 (16 entries) that reported on placental growth factor-based models. Fourteen studies reported on performance of placental growth factor testing for the prediction of preeclampsia in the third trimester, including 10 (with 18 entries) that reported on placental growth factor test alone, 8 (with 12 entries) that reported on soluble fms-like tyrosine kinase-1-placental growth factor ratio, and 7 (with 12 entries) that reported on placental growth factor-based models. For the second trimester, Placental growth factor-based models achieved the highest diagnostic odds ratio for the prediction of early preeclampsia in the total population compared with placental growth factor alone and soluble fms-like tyrosine kinase-1-placental growth factor ratio (placental growth factor-based models, 63.20; 95% confidence interval, 37.62-106.16 vs soluble fms-like tyrosine kinase-1-placental growth factor ratio, 6.96; 95% confidence interval, 1.76-27.61 vs placental growth factor alone, 5.62; 95% confidence interval, 3.04-10.38); placental growth factor-based models had higher diagnostic odds ratio than placental growth factor alone for the identification of any-onset preeclampsia in the unselected population (28.45; 95% confidence interval, 13.52-59.85 vs 7.09; 95% confidence interval, 3.74-13.41). For the third trimester, Placental growth factor-based models achieved prediction for any-onset preeclampsia that was significantly better than that of placental growth factor alone but similar to that of soluble fms-like tyrosine kinase-1-placental growth factor ratio (placental growth factor-based models, 27.12; 95% confidence interval, 21.67-33.94 vs placental growth factor alone, 10.31; 95% confidence interval, 7.41-14.35 vs soluble fms-like tyrosine kinase-1-placental growth factor ratio, 14.94; 95% confidence interval, 9.42-23.70). CONCLUSION Placental growth factor with maternal factors ± other biomarkers determined in the second trimester achieved the best predictive performance for early preeclampsia in the total population. However, in the third trimester, placental growth factor-based models had predictive performance for any-onset preeclampsia that was better than that of placental growth factor alone but similar to that of soluble fms-like tyrosine kinase-1-placental growth factor ratio. Through this meta-analysis, we have identified a large number of very heterogeneous studies. Therefore, there is an urgent need to develop standardized research using the same models that combine serum placental growth factor with maternal factors ± other biomarkers to accurately predict preeclampsia. Identification of patients at risk might be beneficial for intensive monitoring and timing delivery.
Collapse
Affiliation(s)
- Piya Chaemsaithong
- Department of Obstetrics and Gynecology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - María M Gil
- Department of Obstetrics and Gynecology, Hospital Universitario de Torrejón, Torrejón de Ardoz, Madrid, Spain; Faculty of Health Sciences, Universidad Francisco de Vitoria, Madrid, Spain
| | - Noppadol Chaiyasit
- Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Diana Cuenca-Gomez
- Department of Obstetrics and Gynecology, Hospital Universitario de Torrejón, Torrejón de Ardoz, Madrid, Spain
| | - Walter Plasencia
- Department of Obstetrics and Gynecology, Complejo Hospitalario Universitario de Canarias, San Cristóbal de La Laguna, Spain
| | - Valeria Rolle
- Biostatistics and Epidemiology Unit, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Liona C Poon
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital, Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region.
| |
Collapse
|
19
|
Kingdom J, Hutcheon JA, Gordijn SJ, El-Demellawy D, Grynspan D. Placental Pathology and Pregnancy Complications. J Clin Med 2023; 12:5053. [PMID: 37568455 PMCID: PMC10419987 DOI: 10.3390/jcm12155053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/20/2023] [Indexed: 08/13/2023] Open
Abstract
Placental pathology assessment following delivery provides an opportunity to identify the presence and type of disease that can mediate major obstetrical complications, especially in cases where the fetus is growth-restricted, born premature, or stillborn, or if the mother suffers from severe hypertensive morbidities [...].
Collapse
Affiliation(s)
- John Kingdom
- Placenta Program, Maternal-Fetal Medicine Division, Department of Obstetrics & Gynaecology, Mount Sinai Hospital, University of Toronto, 600 University Avenue, Toronto, ON M5G 1X5, Canada;
| | - Jennifer A. Hutcheon
- Department of Obstetrics and Gynaecology, University of British Columbia, 4500 Oak Street, Vancouver, BC V6H 2N1, Canada;
| | - Sanne J. Gordijn
- Department of Obstetrics and Gynaecology, University Medical Center of Groningen, CB20, Hanzeplein 1, 9700 RB Groningen, The Netherlands;
| | - Dina El-Demellawy
- Department of Pathology, Children’s Hospital of Eastern Ontario, Ottawa, ON K1H 8L1, Canada;
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - David Grynspan
- Department of Pathology and Laboratory Medicine, Vernon Jubilee Hospital and University of British Columbia, Vancouver, BC V1T 5L2, Canada
| |
Collapse
|
20
|
Freedman AA, Suresh S, Ernst LM. Patterns of placental pathology associated with preeclampsia. Placenta 2023; 139:85-91. [PMID: 37336159 PMCID: PMC10527086 DOI: 10.1016/j.placenta.2023.06.007] [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: 03/16/2023] [Revised: 05/19/2023] [Accepted: 06/10/2023] [Indexed: 06/21/2023]
Abstract
INTRODUCTION Maternal vascular malperfusion (MVM) is commonly observed in early onset preeclampsia, but less prevalent in late onset preeclampsia. The purpose of our analysis was to investigate patterns of placental pathology in preeclampsia. METHODS Electronic health records for all singleton livebirths from 2009 to 2018 at a single institution with a diagnosis of preeclampsia were obtained. Text searching was used to obtain placental data from pathology reports, including lesions of MVM, fetal vascular malperfusion (FVM), chronic inflammation (CI), and acute inflammation (AI). Placental pathology was compared based on timing of delivery and latent class analysis (LCA) was used to investigate subtypes of preeclampsia based on 22 placental variables. RESULTS 728 patients were included in the analysis. Prevalence of MVM decreased with advancing gestation (95.4% at <34 weeks, 69.8% at 34-36 weeks, and 50%, ≥37 weeks; p < 0.01). LCA identified five classes based on placental pathology: (1) high grade MVM, (2) CI and FVM, (3) low grade MVM, (4) AI, (5) other. Preterm birth varied across the classes (p < 0.01), with the highest prevalence observed among the classes characterized by MVM (high grade: 87.6%; low grade: 63.0%) and the lowest prevalence among the class characterized by AI (23.5%). DISCUSSION Placental pathology in preeclampsia differs based on gestational age at delivery with MVM seen in nearly all early onset preeclampsia cases. Latent classes largely grouped by previously defined patterns of placental injury (MVM, CI, FVM, AI), and again revealed the highest likelihood of preterm birth in classes characterized by MVM. Results suggest there may be multiple mechanisms leading to the clinical manifestations of preeclampsia.
Collapse
Affiliation(s)
- Alexa A Freedman
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, United States.
| | - Sunitha Suresh
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, United States
| | - Linda M Ernst
- Department of Pathology and Laboratory Medicine, NorthShore University HealthSystem, Evanston, IL, United States; Department of Pathology, University of Chicago Pritzker School of Medicine, Chicago, IL, United States
| |
Collapse
|
21
|
Brockway HM, Wilson SL, Kallapur SG, Buhimschi CS, Muglia LJ, Jones HN. Characterization of methylation profiles in spontaneous preterm birth placental villous tissue. PLoS One 2023; 18:e0279991. [PMID: 36952446 PMCID: PMC10035933 DOI: 10.1371/journal.pone.0279991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Indexed: 03/25/2023] Open
Abstract
Preterm birth is a global public health crisis which results in significant neonatal and maternal mortality. Yet little is known regarding the molecular mechanisms of idiopathic spontaneous preterm birth, and we have few diagnostic markers for adequate assessment of placental development and function. Previous studies of placental pathology and our transcriptomics studies suggest a role for placental maturity in idiopathic spontaneous preterm birth. It is known that placental DNA methylation changes over gestation. We hypothesized that if placental hypermaturity is present in our samples, we would observe a unique idiopathic spontaneous preterm birth DNA methylation profile potentially driving the gene expression differences we previously identified in our placental samples. Our results indicate the idiopathic spontaneous preterm birth DNA methylation pattern mimics the term birth methylation pattern suggesting hypermaturity. Only seven significant differentially methylated regions fitting the idiopathic spontaneous preterm birth specific (relative to the controls) profile were identified, indicating unusually high similarity in DNA methylation between idiopathic spontaneous preterm birth and term birth samples. We identified an additional 1,718 significantly methylated regions in our gestational age matched controls where the idiopathic spontaneous preterm birth DNA methylation pattern mimics the term birth methylation pattern, again indicating a striking level of similarity between the idiopathic spontaneous preterm birth and term birth samples. Pathway analysis of these regions revealed differences in genes within the WNT and Cadherin signaling pathways, both of which are essential in placental development and maturation. Taken together, these data demonstrate that the idiopathic spontaneous preterm birth samples display a hypermature methylation signature than expected given their respective gestational age which likely impacts birth timing.
Collapse
Affiliation(s)
- Heather M. Brockway
- Department of Physiology and Functional Genomics, College of Medicine at the University of Florida, Gainesville, Florida, United States of America
| | - Samantha L. Wilson
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Suhas G. Kallapur
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California, UCLA Mattel Children’s Hospital, Los Angeles, California, United States of America
| | - Catalin S. Buhimschi
- Department of Obstetrics and Gynecology, The University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Louis J. Muglia
- Burroughs Wellcome Fund, Research Triangle Park, North Carolina, United States of America
| | - Helen N. Jones
- Department of Physiology and Functional Genomics, College of Medicine at the University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
22
|
Placental Mitochondrial Function and Dysfunction in Preeclampsia. Int J Mol Sci 2023; 24:ijms24044177. [PMID: 36835587 PMCID: PMC9963167 DOI: 10.3390/ijms24044177] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
The placenta is a vital organ of pregnancy, regulating adaptation to pregnancy, gestational parent/fetal exchange, and ultimately, fetal development and growth. Not surprisingly, in cases of placental dysfunction-where aspects of placental development or function become compromised-adverse pregnancy outcomes can result. One common placenta-mediated disorder of pregnancy is preeclampsia (PE), a hypertensive disorder of pregnancy with a highly heterogeneous clinical presentation. The wide array of clinical characteristics observed in pregnant individuals and neonates of a PE pregnancy are likely the result of distinct forms of placental pathology underlying the PE diagnosis, explaining why no one common intervention has proven effective in the prevention or treatment of PE. The historical paradigm of placental pathology in PE highlights an important role for utero-placental malperfusion, placental hypoxia and oxidative stress, and a critical role for placental mitochondrial dysfunction in the pathogenesis and progression of the disease. In the current review, the evidence of placental mitochondrial dysfunction in the context of PE will be summarized, highlighting how altered mitochondrial function may be a common feature across distinct PE subtypes. Further, advances in this field of study and therapeutic targeting of mitochondria as a promising intervention for PE will be discussed.
Collapse
|
23
|
Redline RW, Roberts DJ, Parast MM, Ernst LM, Morgan TK, Greene MF, Gyamfi-Bannerman C, Louis JM, Maltepe E, Mestan KK, Romero R, Stone J. Placental pathology is necessary to understand common pregnancy complications and achieve an improved taxonomy of obstetrical disease. Am J Obstet Gynecol 2023; 228:187-202. [PMID: 35973475 PMCID: PMC10337668 DOI: 10.1016/j.ajog.2022.08.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/08/2022] [Accepted: 08/08/2022] [Indexed: 01/28/2023]
Abstract
The importance of a fully functioning placenta for a good pregnancy outcome is unquestioned. Loss of function can lead to pregnancy complications and is often detected by a thorough placental pathologic examination. Placental pathology has advanced the science and practice of obstetrics and neonatal-perinatal medicine by classifying diseases according to underlying biology and specific patterns of injury. Many past obstacles have limited the incorporation of placental findings into both clinical studies and day-to-day practice. Limitations have included variability in the nomenclature used to describe placental lesions, a shortage of perinatal pathologists fully competent to analyze placental specimens, and a troubling lack of understanding of placental diagnoses by clinicians. However, the potential use of placental pathology for phenotypic classification, improved understanding of the biology of adverse pregnancy outcomes, the development of treatment and prevention, and patient counseling has never been greater. This review, written partly in response to a recent critique published in a major obstetrics-gynecology journal, reexamines the role of placental pathology by reviewing current concepts of biology; explaining the most recent terminology; emphasizing the usefulness of specific diagnoses for obstetrician-gynecologists, neonatologists, and patients; previewing upcoming changes in recommendations for placental submission; and suggesting future improvements. These improvements should include further consideration of overall healthcare costs, cost-effectiveness, the clinical value added of placental assessment, improvements in placental pathology education and practice, and leveraging of placental pathology to identify new biomarkers of disease and evaluate novel therapies tailored to specific clinicopathologic phenotypes of both women and infants.
Collapse
Affiliation(s)
- Raymond W Redline
- Department of Pathology and Reproductive Biology, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center and Cleveland, OH.
| | - Drucilla J Roberts
- Department of Pathology, Harvard Medical School and Massachusetts General Hospital, Boston, MA
| | - Mana M Parast
- Department of Pathology, Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA
| | - Linda M Ernst
- Department of Pathology and Laboratory Medicine, NorthShore University HealthSystem, Evanston, IL
| | - Terry K Morgan
- Department of Pathology and Obstetrics and Gynecology, Center for Developmental Health, Oregon Health Sciences University, Portland, OR
| | - Michael F Greene
- Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School and Massachusetts General Hospital, Boston, MA
| | - Cynthia Gyamfi-Bannerman
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, La Jolla, CA
| | - Judette M Louis
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University South Florida, Tampa, FL
| | - Emin Maltepe
- Department of Pediatrics, University California, San Francisco, San Francisco, CA
| | - Karen K Mestan
- Department of Pediatrics and Neonatology, University of California, San Diego, School of Medicine and Rady Children's Hospital, San Diego, CA
| | - 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, Maryland, 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
| | - Joanne Stone
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
24
|
Horii M, To C, Morey R, Jacobs MB, Li Y, Nelson KK, Meads M, Siegel BA, Pizzo D, Adami R, Zhang-Rutledge K, Lamale-Smith L, Laurent LC, Parast MM. Histopathologic and Transcriptomic Profiling Identifies Novel Trophoblast Defects in Patients With Preeclampsia and Maternal Vascular Malperfusion. Mod Pathol 2023; 36:100035. [PMID: 36853788 PMCID: PMC10081686 DOI: 10.1016/j.modpat.2022.100035] [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: 04/29/2022] [Revised: 08/03/2022] [Accepted: 09/28/2022] [Indexed: 01/11/2023]
Abstract
Preeclampsia (PE) is a heterogeneous disease for which the current clinical classification system is based on the presence or absence of specific clinical features. PE-associated placentas also show heterogeneous findings on pathologic examination, suggesting that further subclassification is possible. We combined clinical, pathologic, immunohistochemical, and transcriptomic profiling of placentas to develop integrated signatures for multiple subclasses of PE. In total, 303 PE and 1388 nonhypertensive control placentas were included. We found that maternal vascular malperfusion (MVM) in the placenta was associated with preterm PE with severe features and with small-for-gestational-age neonates. Interestingly, PE placentas with either MVM or no histologic pattern of injury showed a linear decrease in proliferative (p63+) cytotrophoblast per villous area with increasing gestational age, similar to placentas obtained from the nonhypertensive patient cohort; however, PE placentas with fetal vascular malperfusion or villitis of unknown etiology lost this phenotype. This is mainly because of cases of fetal vascular malperfusion in placentas of patients with preterm PE and villitis of unknown etiology in placentas of patients with term PE, which are associated with a decrease or increase, respectively, in the cytotrophoblast per villous area. Finally, a transcriptomic analysis identified pathways associated with hypoxia, inflammation, and reduced cell proliferation in PE-MVM placentas and further subclassified this group into extravillous trophoblast-high and extravillous trophoblast-low PE, confirmed using an immunohistochemical analysis of trophoblast lineage-specific markers. Our findings suggest that within specific histopathologic patterns of placental injury, PE can be subclassified based on specific cellular and molecular defects, allowing the identification of pathways that may be targeted for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Mariko Horii
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Cuong To
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California
| | - Robert Morey
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California
| | - Marni B Jacobs
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California
| | - Yingchun Li
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Katharine K Nelson
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Morgan Meads
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Brent A Siegel
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Donald Pizzo
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Rebecca Adami
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California
| | - Kathy Zhang-Rutledge
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California
| | - Leah Lamale-Smith
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California
| | - Louise C Laurent
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California.
| |
Collapse
|
25
|
Wang Z, Liu D, Dai Y, Li R, Zheng Y, Zhao G, Wang J, Diao Z, Cao C, Lv H, Gu N, Zhou H, Ding H, Li J, Zhu X, Duan H, Shen L, Zhang Q, Chen J, Hu H, Wang X, Zheng M, Zhou Y, Hu Y. Elevated Placental microRNA-155 Is a Biomarker of a Preeclamptic Subtype. Hypertension 2023; 80:370-384. [PMID: 36519433 DOI: 10.1161/hypertensionaha.122.19914] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Preeclampsia is a complicated syndrome with marked heterogeneity. The biomarker-based classification for this syndrome is more constructive to the targeted prevention and treatment of preeclampsia. It has been reported that preeclamptic patients had elevated microRNA-155 (miR-155) in placentas or circulation. Here, we investigated the characteristics of patients with high placental miR-155 (pl-miR-155). METHODS Based on the 95th percentile (P95) of pl-miR-155 in controls, preeclamptic patients were divided into high miR-155 group (≥P95) and normal miR-155 group (<P95). The changes of placental pathology, clinical manifestations, and placental transcriptome of preeclamptic patients were clustered by t-distributed stochastic neighbor embedding and hierarchical clustering analysis. The placental restricted miR-155 overexpression mouse model was constructed, and the phenotype, placental pathology, and transcriptome were evaluated. Furthermore, the therapeutic potential of antagonist of miR-155 was explored by administrating with antagomir-155. RESULTS About one-third of preeclamptic patients had high pl-miR-155 expression, which was positively correlated with circulating miR-155 levels. These patients could be clustered as 1 group, according to clinical manifestation, placental pathology, or transcriptomes by t-distributed stochastic neighbor embedding and hierarchical clustering analysis. Further, the pregnant mice with placental restricted miR-155 overexpression could simulate the changes of clinical signs, pathology, and transcriptome of placentas in patients with high pl-miR-155. AntagomiR-155 treatment relieved the preeclampsia-like phenotype and improved the placental vascular development in mice. CONCLUSIONS There is at least 1 type of preeclampsia with upregulated miR-155 presenting more severe clinical manifestations. MiR-155 may be a potential therapeutic target in patients with high pl-miR-155.
Collapse
Affiliation(s)
- Zhiyin Wang
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Dan Liu
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Yimin Dai
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Ruotian Li
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (R.L.)
| | - Yaowu Zheng
- Transgenic Research Center, Northeast Normal University, Changchun, China (Y.Z.)
| | - Guangfeng Zhao
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Jingmei Wang
- Department of Pathology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (J.W.)
| | - Zhenyu Diao
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Chenrui Cao
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Haining Lv
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Ning Gu
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Hang Zhou
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Hailin Ding
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Jie Li
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Xiangyu Zhu
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Honglei Duan
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Li Shen
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Qun Zhang
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Jing Chen
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Huilian Hu
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Xiaoyan Wang
- The Core Laboratory for Clinical Research, The Affiliated BenQ Hospital of Nanjing Medical University, China (X.W.)
| | - Mingming Zheng
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| | - Yan Zhou
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco (Y.Z.)
| | - Yali Hu
- From the Center for Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China (Z.W., D.L., Y.D., G.Z., Z.D., C.C., H.L., N.G., H.Z., H.D., J.L., X.Z., H.D., L.S., Q.Z., J.C., H.H., M.Z., Y.H.)
| |
Collapse
|
26
|
The imbalance of circulating monocyte subgroups with a higher proportion of the CD14+CD16+CD163+ phenotype in patients with preeclampsia. Immunol Lett 2023; 253:1-7. [PMID: 36460232 DOI: 10.1016/j.imlet.2022.11.005] [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: 09/08/2022] [Revised: 11/20/2022] [Accepted: 11/27/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Preeclampsia is a major cause of increased maternal and fetal morbidity and mortality, which is closely related to the abnormal maternal immune response. The skew of decidual macrophage polarization toward M1 phenotype has been proved to promote the pathogenesis of preeclampsia. However, it's not easy to monitor the change of decidual macrophage subtypes. The current study aims to examine the distribution of different circulating monocyte subtypes and analyze whether certain monocyte subtypes act as potential clinical indicators for preeclampsia. METHODS A total of 50 pregnant women [mild preeclampsia (n = 20); severe preeclampsia (n = 15); healthy pregnancy (n = 15)] and 15 healthy donors were included in the study. Medical information such as BMI, blood pressure, ALT, creatinine, thrombocyte, etc., were recorded. The frequency of different monocyte subtypes in venous blood were measured by flow cytometry. Serum level of IL-6 was detected using Roche-Hitachi cobas 8000. Serum concentration of inflammatory cytokines (IL-1β, IL-4, IL-10 and TNF-α) were measured by ELISA. RESULTS A circulating monocyte subset with both M1 and M2 markers (CD14+CD16+CD163+) was found to occupy an obvious higher proportion in the preeclampsia group than in the normal pregnancy group. The ratio of CD206+/CD206- M2-like monocytes was also increased in the preeclampsia group, and meanwhile, it had statistic difference between the mild- and the severe-preeclampsia group. Furthermore, the serum levels of IL-1β and TNF-α were positively correlated with the frequency of CD14+CD16+CD163+ intermediate monocytes in the preeclampsia group. CONCLUSIONS The increased proportion of CD14+C16+CD163+ circulating monocytes and the high ratio of CD206+/CD206- M2-like monocytes may act as potential clinical indicators for preeclampsia, with the superiority of convenience and dynamic monitoring.
Collapse
|
27
|
Kedziora SM, Obermayer B, Sugulle M, Herse F, Kräker K, Haase N, Langmia IM, Müller DN, Staff AC, Beule D, Dechend R. Placental Transcriptome Profiling in Subtypes of Diabetic Pregnancies Is Strongly Confounded by Fetal Sex. Int J Mol Sci 2022; 23:ijms232315388. [PMID: 36499721 PMCID: PMC9740420 DOI: 10.3390/ijms232315388] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
The placenta is a temporary organ with a unique structure and function to ensure healthy fetal development. Placental dysfunction is involved in pre-eclampsia (PE), fetal growth restriction, preterm birth, and gestational diabetes mellitus (GDM). A diabetic state affects maternal and fetal health and may lead to functional alterations of placental metabolism, inflammation, hypoxia, and weight, amplifying the fetal stress. The placental molecular adaptations to the diabetic environment and the adaptive spatio-temporal consequences to elevated glucose or insulin are largely unknown (2). We aimed to identify gene expression signatures related to the diabetic placental pathology of placentas from women with diabetes mellitus. Human placenta samples (n = 77) consisting of healthy controls, women with either gestational diabetes mellitus (GDM), type 1 or type 2 diabetes, and women with GDM, type 1 or type 2 diabetes and superimposed PE were collected. Interestingly, gene expression differences quantified by total RNA sequencing were mainly driven by fetal sex rather than clinical diagnosis. Association of the principal components with a full set of clinical patient data identified fetal sex as the single main explanatory variable. Accordingly, placentas complicated by type 1 and type 2 diabetes showed only few differentially expressed genes, while possible effects of GDM and diabetic pregnancy complicated by PE were not identifiable in this cohort. We conclude that fetal sex has a prominent effect on the placental transcriptome, dominating and confounding gene expression signatures resulting from diabetes mellitus in settings of well-controlled diabetic disease. Our results support the notion of placenta as a sexual dimorphic organ.
Collapse
Affiliation(s)
- Sarah M. Kedziora
- Experimental and Clinical Research Center (ECRC), a Joint Cooperation between the Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, 10178 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, 10785 Berlin, Germany
| | - Benedikt Obermayer
- Berlin Institute of Health, Charité—Universitätsmedizin Berlin, Core Unit Bioinformatics, 10117 Berlin, Germany
| | - Meryam Sugulle
- Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Division of Obstetrics and Gynaecology, Oslo University Hospital, 0424 Oslo, Norway
| | - Florian Herse
- Experimental and Clinical Research Center (ECRC), a Joint Cooperation between the Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, 10178 Berlin, Germany
| | - Kristin Kräker
- Experimental and Clinical Research Center (ECRC), a Joint Cooperation between the Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, 10178 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, 10785 Berlin, Germany
| | - Nadine Haase
- Experimental and Clinical Research Center (ECRC), a Joint Cooperation between the Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, 10178 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, 10785 Berlin, Germany
| | - Immaculate M. Langmia
- Experimental and Clinical Research Center (ECRC), a Joint Cooperation between the Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, 10178 Berlin, Germany
| | - Dominik N. Müller
- Experimental and Clinical Research Center (ECRC), a Joint Cooperation between the Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, 10178 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, 10785 Berlin, Germany
| | - Anne Cathrine Staff
- Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Division of Obstetrics and Gynaecology, Oslo University Hospital, 0424 Oslo, Norway
| | - Dieter Beule
- Berlin Institute of Health, Charité—Universitätsmedizin Berlin, Core Unit Bioinformatics, 10117 Berlin, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center (ECRC), a Joint Cooperation between the Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, 10785 Berlin, Germany
- HELIOS Clinic, Department of Cardiology and Nephrology, 13125 Berlin, Germany
- Correspondence: ; Tel.: +49-30-4505-40301
| |
Collapse
|
28
|
Wang LQ, Fernandez-Boyano I, Robinson WP. Genetic variation in placental insufficiency: What have we learned over time? Front Cell Dev Biol 2022; 10:1038358. [PMID: 36313546 PMCID: PMC9613937 DOI: 10.3389/fcell.2022.1038358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/03/2022] [Indexed: 11/28/2022] Open
Abstract
Genetic variation shapes placental development and function, which has long been known to impact fetal growth and pregnancy outcomes such as miscarriage or maternal pre-eclampsia. Early epidemiology studies provided evidence of a strong heritable component to these conditions with both maternal and fetal-placental genetic factors contributing. Subsequently, cytogenetic studies of the placenta and the advent of prenatal diagnosis to detect chromosomal abnormalities provided direct evidence of the importance of spontaneously arising genetic variation in the placenta, such as trisomy and uniparental disomy, drawing inferences that remain relevant to this day. Candidate gene approaches highlighted the role of genetic variation in genes influencing immune interactions at the maternal-fetal interface and angiogenic factors. More recently, the emergence of molecular techniques and in particular high-throughput technologies such as Single-Nucleotide Polymorphism (SNP) arrays, has facilitated the discovery of copy number variation and study of SNP associations with conditions related to placental insufficiency. This review integrates past and more recent knowledge to provide important insights into the role of placental function on fetal and perinatal health, as well as into the mechanisms leading to genetic variation during development.
Collapse
Affiliation(s)
- Li Qing Wang
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Icíar Fernandez-Boyano
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Wendy P. Robinson
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
29
|
Romero R, Jung E, Chaiworapongsa T, Erez O, Gudicha DW, Kim YM, Kim JS, Kim B, Kusanovic JP, Gotsch F, Taran AB, Yoon BH, Hassan SS, Hsu CD, Chaemsaithong P, Gomez-Lopez N, Yeo L, Kim CJ, Tarca AL. Toward a new taxonomy of obstetrical disease: improved performance of maternal blood biomarkers for the great obstetrical syndromes when classified according to placental pathology. Am J Obstet Gynecol 2022; 227:615.e1-615.e25. [PMID: 36180175 PMCID: PMC9525890 DOI: 10.1016/j.ajog.2022.04.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND The major challenge for obstetrics is the prediction and prevention of the great obstetrical syndromes. We propose that defining obstetrical diseases by the combination of clinical presentation and disease mechanisms as inferred by placental pathology will aid in the discovery of biomarkers and add specificity to those already known. OBJECTIVE To describe the longitudinal profile of placental growth factor (PlGF), soluble fms-like tyrosine kinase-1 (sFlt-1), and the PlGF/sFlt-1 ratio throughout gestation, and to determine whether the association between abnormal biomarker profiles and obstetrical syndromes is strengthened by information derived from placental examination, eg, the presence or absence of placental lesions of maternal vascular malperfusion. STUDY DESIGN This retrospective case cohort study was based on a parent cohort of 4006 pregnant women enrolled prospectively. The case cohort of 1499 pregnant women included 1000 randomly selected patients from the parent cohort and all additional patients with obstetrical syndromes from the parent cohort. Pregnant women were classified into six groups: 1) term delivery without pregnancy complications (n=540; control); 2) preterm labor and delivery (n=203); 3) preterm premature rupture of the membranes (n=112); 4) preeclampsia (n=230); 5) small-for-gestational-age neonate (n=334); and 6) other pregnancy complications (n=182). Maternal plasma concentrations of PlGF and sFlt-1 were determined by enzyme-linked immunosorbent assays in 7560 longitudinal samples. Placental pathologists, masked to clinical outcomes, diagnosed the presence or absence of placental lesions of maternal vascular malperfusion. Comparisons between mean biomarker concentrations in cases and controls were performed by utilizing longitudinal generalized additive models. Comparisons were made between controls and each obstetrical syndrome with and without subclassifying cases according to the presence or absence of placental lesions of maternal vascular malperfusion. RESULTS 1) When obstetrical syndromes are classified based on the presence or absence of placental lesions of maternal vascular malperfusion, significant differences in the mean plasma concentrations of PlGF, sFlt-1, and the PlGF/sFlt-1 ratio between cases and controls emerge earlier in gestation; 2) the strength of association between an abnormal PlGF/sFlt-1 ratio and the occurrence of obstetrical syndromes increases when placental lesions of maternal vascular malperfusion are present (adjusted odds ratio [aOR], 13.6 vs 6.7 for preeclampsia; aOR, 8.1 vs 4.4 for small-for-gestational-age neonates; aOR, 5.5 vs 2.1 for preterm premature rupture of the membranes; and aOR, 3.3 vs 2.1 for preterm labor (all P<0.05); and 3) the PlGF/sFlt-1 ratio at 28 to 32 weeks of gestation is abnormal in patients who subsequently delivered due to preterm labor with intact membranes and in those with preterm premature rupture of the membranes if both groups have placental lesions of maternal vascular malperfusion. Such association is not significant in patients with these obstetrical syndromes who do not have placental lesions. CONCLUSION Classification of obstetrical syndromes according to the presence or absence of placental lesions of maternal vascular malperfusion allows biomarkers to be informative earlier in gestation and enhances the strength of association between biomarkers and clinical outcomes. We propose that a new taxonomy of obstetrical disorders informed by placental pathology will facilitate the discovery and implementation of biomarkers as well as the prediction and prevention of such disorders.
Collapse
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 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.
| | - Eunjung Jung
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Offer Erez
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Faculty of Health Sciences, Division of Obstetrics and Gynecology, Maternity Department "D," Soroka University Medical Center, School of Medicine, Ben-Gurion University of the Negev, Beersheba, Israel; Department of Obstetrics and Gynecology, HaEmek Medical Center, Afula, Israel
| | - Dereje W Gudicha
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Yeon Mee Kim
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Pathology, Wayne State University School of Medicine, Detroit, MI; Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Jung-Sun Kim
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Pathology, Wayne State University School of Medicine, Detroit, MI; Department of Pathology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Bomi Kim
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Pathology, Wayne State University School of Medicine, Detroit, MI; Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Juan Pedro Kusanovic
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; División de Obstetricia y Ginecología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro de Investigación e Innovación en Medicina Materno-Fetal, Unidad de Alto Riesgo Obstétrico, Hospital Sotero Del Rio, Santiago, Chile
| | - Francesca Gotsch
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Andreea B Taran
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Bo Hyun Yoon
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sonia S Hassan
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Office of Women's Health, Integrative Biosciences Center, Wayne State University, Detroit, MI; Department of Physiology, Wayne State University School of Medicine, Detroit, MI
| | - Chaur-Dong Hsu
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Physiology, Wayne State University School of Medicine, Detroit, MI; Department of Obstetrics and Gynecology, University of Arizona, College of Medicine - Tucson, Tucson, AZ
| | - Piya Chaemsaithong
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Faculty of Medicine, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI
| | - Lami Yeo
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Chong Jai Kim
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Pathology, Wayne State University School of Medicine, Detroit, MI; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Adi L Tarca
- Perinatology Research Branch, Divisions of Obstetrics and Maternal-Fetal Medicine and Intramural Research, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Computer Science, Wayne State University College of Engineering, Detroit, MI
| |
Collapse
|
30
|
Maternal Immune Cell and Cytokine Profiles to Predict Cardiovascular Risk Six Months after Preeclampsia. J Clin Med 2022; 11:jcm11144185. [PMID: 35887949 PMCID: PMC9317739 DOI: 10.3390/jcm11144185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/13/2022] [Accepted: 07/16/2022] [Indexed: 11/20/2022] Open
Abstract
Women who develop preeclampsia (PE) are at high risk for cardiovascular disease (CVD). Early identification of women with PE who may benefit the most from early cardiovascular risk screening and interventions remains challenging. Our objective was to assess whether cytokine and immune cell profiles after PE are helpful in distinguishing women at low and high CVD risk at 6-months postpartum. Individuals who developed PE were followed for immune cell phenotyping and plasma cytokine quantification at delivery, at 3-months, and at 6-months postpartum. Lifetime CVD risk was assessed at 6-months postpartum, and the immune cell and cytokine profiles were compared between risk groups at each time point. Among 31 participants, 18 (58.1%) exhibited high CVD-risk profiles at 6-months postpartum. The proportion of circulating NK-cells was significantly lower in high-risk participants at delivery (p = 0.04). At 3-months postpartum, high-risk participants exhibited a lower proportion of FoxP3+ regulatory T-cells (p = 0.01), a greater proportion of CD8+ T cells (p = 0.02) and a lower CD4+:CD8+ ratio (p = 0.02). There were no differences in immune cell populations at 6-months postpartum. There were no differences in plasma cytokines levels between risk groups at any time point. Subtle differences in immune cell profiles may help distinguish individuals at low and high CVD risk in the early postpartum period and warrants further investigation.
Collapse
|
31
|
Barr LC, Liblik K, Johri AM, Smith GN. Maternal Cardiovascular Function Following a Pregnancy Complicated by Preeclampsia. Am J Perinatol 2022; 39:1055-1064. [PMID: 33321533 DOI: 10.1055/s-0040-1721694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Preeclampsia is a hypertensive pregnancy complication with an unknown etiology and high maternal burden worldwide. Burgeoning research has linked preeclampsia to adverse maternal health outcomes remote from pregnancy; however, the intermediary mechanisms responsible for this association have not been sufficiently established. In the present narrative review, we summarize leading evidence of structural and functional cardiovascular changes associated with prior preeclampsia, and how these changes may be linked to future maternal disease. KEY POINTS: · Prior preeclampsia is associated with subclinical structural and functional vascular changes remote from pregnancy.. · Maternal cardiac adaptations to preeclampsia may have long-term implications on cardiovascular health.. · Clinicians have an opportunity to minimize maternal disease risk following preeclampsia..
Collapse
Affiliation(s)
- Logan C Barr
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Kiera Liblik
- Cardiovascular Imaging Network at Queen's, Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Amer M Johri
- Cardiovascular Imaging Network at Queen's, Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Graeme N Smith
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
32
|
Jaiman S, Romero R, Bhatti G, Jung E, Gotsch F, Suksai M, Gallo DM, Chaiworapongsa T, Kadar N. The role of the placenta in spontaneous preterm labor and delivery with intact membranes. J Perinat Med 2022; 50:553-566. [PMID: 35246973 PMCID: PMC9189066 DOI: 10.1515/jpm-2021-0681] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/20/2022] [Indexed: 12/16/2022]
Abstract
OBJECTIVES To determine whether placental vascular pathology and impaired placental exchange due to maturational defects are involved in the etiology of spontaneous preterm labor and delivery in cases without histologic acute chorioamnionitis. METHODS This was a retrospective, observational study. Cases included pregnancies that resulted in spontaneous preterm labor and delivery (<37 weeks), whereas uncomplicated pregnancies that delivered fetuses at term (≥37-42 weeks of gestation) were selected as controls. Placental histological diagnoses were classified into three groups: lesions of maternal vascular malperfusion, lesions of fetal vascular malperfusion, and placental microvasculopathy, and the frequency of each type of lesion in cases and controls was compared. Moreover, we specifically searched for villous maturational abnormalities in cases and controls. Doppler velocimetry of the umbilical and uterine arteries were performed in a subset of patients. RESULTS There were 184 cases and 2471 controls, of which 95 and 1178 had Doppler studies, respectively. The frequency of lesions of maternal vascular malperfusion was greater in the placentas of patients with preterm labor than in the control group [14.1% (26/184) vs. 8.8% (217/2471) (p=0.023)]. Disorders of villous maturation were more frequent in the group with preterm labor than in the control group: 41.1% (39/95) [delayed villous maturation in 31.6% (30/95) vs. 2.5% (13/519) in controls and accelerated villous maturation in 9.5% (9/95) vs. none in controls]. CONCLUSIONS Maturational defects of placental villi were associated with approximately 41% of cases of unexplained spontaneous preterm labor and delivery without acute inflammatory lesions of the placenta and with delivery of appropriate-for-gestational-age fetuses.
Collapse
Affiliation(s)
- Sunil Jaiman
- 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, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), 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
| | - 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, Maryland, and Detroit, Michigan, USA
- Department of Biomedical Engineering, Wayne State University College of Engineering, Detroit, MI, 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 (NICHD/NIH/DHHS), 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 (NICHD/NIH/DHHS), 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 (NICHD/NIH/DHHS), 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 (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - 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 (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | |
Collapse
|
33
|
Fakhr Y, Koshti S, Habibyan YB, Webster K, Hemmings DG. Tumor Necrosis Factor-α Induces a Preeclamptic-like Phenotype in Placental Villi via Sphingosine Kinase 1 Activation. Int J Mol Sci 2022; 23:ijms23073750. [PMID: 35409108 PMCID: PMC8998215 DOI: 10.3390/ijms23073750] [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: 02/03/2022] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 02/01/2023] Open
Abstract
Preeclampsia (PE) involves inadequate placental function. This can occur due to elevated pro-inflammatory tumor necrosis factor-α (TNF-α). In other tissues, TNF-α signals via sphingosine kinase 1 (SphK1). SphK1 hinders syncytial formation. Whether this occurs downstream of TNF-α signaling is unclear. We hypothesized that placental SphK1 levels are higher in PE and elevated TNF-α decreases syncytial function, increases syncytial shedding, and increases cytokine/factor release via SphK1 activity. Term placental biopsies were analyzed for SphK1 using immunofluorescence and qRT-PCR. Term placental explants were treated after 4 days of culture, at the start of syncytial regeneration, with TNF-α and/or SphK1 inhibitors, PF-543. Syncytialization was assessed by measuring fusion and chorionic gonadotropin release. Cell death and shedding were measured by lactate dehydrogenase release and placental alkaline phosphatase-positive shed particles. Forty-two cytokines were measured using multiplex assays. Placental SphK1 was increased in PE. Increased cell death, shedding, interferon-α2, IFN-γ-induced protein 10, fibroblast growth factor 2, and platelet-derived growth factor-AA release induced by TNF-α were reversed upon SphK1 inhibition. TNF-α increased the release of 26 cytokines independently of SphK1. TNF-α decreased IL-10 release and inhibiting SphK1 reversed this effect. Inhibiting SphK1 alone decreased TNF-α release. Hence, SphK1 partially mediates the TNF-α-induced PE placental phenotype, primarily through cell damage, shedding, and specific cytokine release.
Collapse
Affiliation(s)
- Yuliya Fakhr
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T5H 3V9, Canada; (Y.F.); (S.K.); (Y.B.H.); (K.W.)
- Women and Children’s Health Research Institute, Edmonton, AB T6G 1C9, Canada
| | - Saloni Koshti
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T5H 3V9, Canada; (Y.F.); (S.K.); (Y.B.H.); (K.W.)
- Women and Children’s Health Research Institute, Edmonton, AB T6G 1C9, Canada
| | - Yasaman Bahojb Habibyan
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T5H 3V9, Canada; (Y.F.); (S.K.); (Y.B.H.); (K.W.)
- Women and Children’s Health Research Institute, Edmonton, AB T6G 1C9, Canada
| | - Kirsten Webster
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T5H 3V9, Canada; (Y.F.); (S.K.); (Y.B.H.); (K.W.)
- Women and Children’s Health Research Institute, Edmonton, AB T6G 1C9, Canada
| | - Denise G. Hemmings
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T5H 3V9, Canada; (Y.F.); (S.K.); (Y.B.H.); (K.W.)
- Women and Children’s Health Research Institute, Edmonton, AB T6G 1C9, Canada
- Department of Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Correspondence: ; Tel.: +1-(780)-492-2098
| |
Collapse
|
34
|
Benton SJ, Mery EE, Grynspan D, Gaudet LM, Smith GN, Bainbridge SA. Placental Pathology as a Tool to Identify Women for Postpartum Cardiovascular Risk Screening following Preeclampsia: A Preliminary Investigation. J Clin Med 2022; 11:jcm11061576. [PMID: 35329905 PMCID: PMC8954716 DOI: 10.3390/jcm11061576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/03/2022] [Accepted: 03/11/2022] [Indexed: 12/11/2022] Open
Abstract
Preeclampsia (PE) is associated with an increased risk of cardiovascular disease (CVD) in later life. Postpartum cardiovascular risk screening could identify patients who would benefit most from early intervention and lifestyle modification. However, there are no readily available methods to identify these high-risk women. We propose that placental lesions may be useful in this regard. Here, we determine the association between placental lesions and lifetime CVD risk assessed 6 months following PE. Placentas from 85 PE women were evaluated for histopathological lesions. At 6 months postpartum, a lifetime cardiovascular risk score was calculated. Placental lesions were compared between CVD risk groups and the association was assessed using odds ratios. Multivariable logistic regression was used to develop prediction models for CVD risk with placental pathology. Placentas from high-risk women had more severe lesions of maternal vascular malperfusion (MVM) and resulted in a 3-fold increased risk of screening as high-risk for CVD (OR 3.10 (1.20–7.92)) compared to women without these lesions. MVM lesion severity was moderately predictive of high-risk screening (AUC 0.63 (0.51, 0.75); sensitivity 71.8% (54.6, 84.4); specificity 54.7% (41.5, 67.3)). When clinical parameters were added, the model’s predictive performance improved (AUC 0.73 (0.62, 0.84); sensitivity 78.4% (65.4, 87.5); specificity 51.6% (34.8, 68.0)). The results suggest that placenta pathology may provide a unique modality to identify women for cardiovascular screening.
Collapse
Affiliation(s)
- Samantha J. Benton
- Department of Health Sciences, Carleton University, Ottawa, ON K1S 5B6, Canada;
| | - Erika E. Mery
- School of Interdisciplinary Health Sciences, University of Ottawa, Ottawa, ON K1H 8L1, Canada;
| | - David Grynspan
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada;
| | - Laura M. Gaudet
- Department of Obstetrics and Gynaecology, Queen’s University, Kingston, ON K7L 2V7, Canada; (L.M.G.); (G.N.S.)
| | - Graeme N. Smith
- Department of Obstetrics and Gynaecology, Queen’s University, Kingston, ON K7L 2V7, Canada; (L.M.G.); (G.N.S.)
| | - Shannon A. Bainbridge
- School of Interdisciplinary Health Sciences, University of Ottawa, Ottawa, ON K1H 8L1, Canada;
- Correspondence:
| |
Collapse
|
35
|
Than NG, Posta M, Györffy D, Orosz L, Orosz G, Rossi SW, Ambrus-Aikelin G, Szilágyi A, Nagy S, Hupuczi P, Török O, Tarca AL, Erez O, Papp Z, Romero R. Early pathways, biomarkers and four distinct molecular subclasses of preeclampsia: The intersection of clinical, pathological and high dimensional biology studies. Placenta 2022; 125:10-19. [DOI: 10.1016/j.placenta.2022.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/18/2022] [Accepted: 03/08/2022] [Indexed: 01/08/2023]
|
36
|
Siddique N, Cox B. Computational analysis identified accelerated senescence as a significant contribution to preeclampsia pathophysiology. Placenta 2022; 121:70-78. [DOI: 10.1016/j.placenta.2022.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/23/2022] [Accepted: 03/03/2022] [Indexed: 01/08/2023]
|
37
|
Sethuraman V, Pu Y, Gingrich J, Jing J, Long R, Olomu IN, Veiga-Lopez A. Expression of ABC transporters during syncytialization in preeclampsia. Pregnancy Hypertens 2022; 27:181-188. [PMID: 35124425 PMCID: PMC9017055 DOI: 10.1016/j.preghy.2022.01.006] [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: 11/24/2020] [Revised: 12/29/2021] [Accepted: 01/23/2022] [Indexed: 01/12/2023]
Abstract
Preeclampsia complicates 2-8% of pregnancies and is associated with prematurity and intrauterine growth restriction. Cholesterol and sterol transport is a key function of the placenta and it is elicited through ATP binding cassette (ABC) transporters. ABCA1 expression changes during trophoblast cell fusion, a process required to form the placental syncytium that enables maternal-fetal nutrient transfer. ABCA1 expression is dysregulated in preeclamptic placentas. But whether ABC transporters expression during trophoblast fusion is disrupted in preeclampsia remains unknown. We investigated if cholesterol and sterol ABC transporters are altered in term and preterm preeclampsia placentas and during human cytotrophoblast syncytialization. Human placental biopsies were collected from healthy term (≥37 weeks; n = 11) and term preeclamptic (≥36 6/7 weeks; n = 8) and pre-term preeclamptic (28-35 weeks; n = 8) pregnancies. Both, protein and mRNA expression for ABCA1, ABCG1, ABCG5, and ABCG8 were evaluated. Primary cytotrophoblasts isolated from a subset of placentas were induced to syncytialize for 96 h and ABCA1, ABCG1 and ABCG8 mRNA expression evaluated at 0 h and 96 h. Protein and gene expression of ABC transporters were not altered in preeclamptic placentas. In the healthy Term group, ABCA1 expression was similar before and after syncytialization. After 96 h of syncytialization, mRNA expression of ABCA1 and ABCG1 increased significantly, while ABCG8 decreased significantly in term-preeclampsia, but not pre-term preeclampsia. While placental expression of ABCA1 and ABCG1 remained unaltered in term preeclampsia, the disruption in their dynamic expression pattern during cytotrophoblast syncytialization suggests that cholesterol transport may contribute to the pathophysiologic role of the placenta in preeclampsia.
Collapse
Affiliation(s)
- Visalakshi Sethuraman
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Yong Pu
- Department of Pathology, University of Illinois at Chicago
| | - Jeremy Gingrich
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, Michigan, USA
| | - Jiongjie Jing
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, Michigan, USA
| | - Robert Long
- Department of Obstetrics and Gynecology, Sparrow Health System, East Lansing, Michigan, USA
| | - Isoken Nicholas Olomu
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Almudena Veiga-Lopez
- Department of Pathology, University of Illinois at Chicago, USA; Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
38
|
Moufarrej MN, Vorperian SK, Wong RJ, Campos AA, Quaintance CC, Sit RV, Tan M, Detweiler AM, Mekonen H, Neff NF, Baruch-Gravett C, Litch JA, Druzin ML, Winn VD, Shaw GM, Stevenson DK, Quake SR. Early prediction of preeclampsia in pregnancy with cell-free RNA. Nature 2022; 602:689-694. [PMID: 35140405 PMCID: PMC8971130 DOI: 10.1038/s41586-022-04410-z] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 01/06/2022] [Indexed: 12/30/2022]
Abstract
Liquid biopsies that measure circulating cell-free RNA (cfRNA) offer an opportunity to study the development of pregnancy-related complications in a non-invasive manner and to bridge gaps in clinical care1-4. Here we used 404 blood samples from 199 pregnant mothers to identify and validate cfRNA transcriptomic changes that are associated with preeclampsia, a multi-organ syndrome that is the second largest cause of maternal death globally5. We find that changes in cfRNA gene expression between normotensive and preeclamptic mothers are marked and stable early in gestation, well before the onset of symptoms. These changes are enriched for genes specific to neuromuscular, endothelial and immune cell types and tissues that reflect key aspects of preeclampsia physiology6-9, suggest new hypotheses for disease progression and correlate with maternal organ health. This enabled the identification and independent validation of a panel of 18 genes that when measured between 5 and 16 weeks of gestation can form the basis of a liquid biopsy test that would identify mothers at risk of preeclampsia long before clinical symptoms manifest themselves. Tests based on these observations could help predict and manage who is at risk for preeclampsia-an important objective for obstetric care10,11.
Collapse
Affiliation(s)
- Mira N Moufarrej
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Sevahn K Vorperian
- ChEM-H and Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Ronald J Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Ana A Campos
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Cecele C Quaintance
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Rene V Sit
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | | | | | | | | | - James A Litch
- Global Alliance to Prevent Prematurity and Stillbirth (GAPPS), Lynnwood, WA, USA
| | - Maurice L Druzin
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Gary M Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - David K Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Stephen R Quake
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
- Department of Applied Physics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
39
|
Pietsch M, Ho A, Bardanzellu A, Zeidan AMA, Chappell LC, Hajnal JV, Rutherford M, Hutter J. APPLAUSE: Automatic Prediction of PLAcental health via U-net Segmentation and statistical Evaluation. Med Image Anal 2021; 72:102145. [PMID: 34229190 PMCID: PMC8350147 DOI: 10.1016/j.media.2021.102145] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 04/26/2021] [Accepted: 06/21/2021] [Indexed: 02/04/2023]
Abstract
PURPOSE Artificial-intelligence population-based automated quantification of placental maturation and health from a rapid functional Magnetic Resonance scan. The placenta plays a crucial role for any successful human pregnancy. Deviations from the normal dynamic maturation throughout gestation are closely linked to major pregnancy complications. Antenatal assessment in-vivo using T2* relaxometry has shown great promise to inform management and possible interventions but clinical translation is hampered by time consuming manual segmentation and analysis techniques based on comparison against normative curves over gestation. METHODS This study proposes a fully automatic pipeline to predict the biological age and health of the placenta based on a free-breathing rapid (sub-30 second) T2* scan in two steps: Automatic segmentation using a U-Net and a Gaussian process regression model to characterize placental maturation and health. These are trained and evaluated on 108 3T MRI placental data sets, the evaluation included 20 high-risk pregnancies diagnosed with pre-eclampsia and/or fetal growth restriction. An independent cohort imaged at 1.5 T is used to assess the generalization of the training and evaluation pipeline. RESULTS Across low- and high-risk groups, automatic segmentation performs worse than inter-rater performance (mean Dice coefficients of 0.58 and 0.68, respectively) but is sufficient for estimating placental mean T2* (0.986 Pearson Correlation Coefficient). The placental health prediction achieves an excellent ability to differentiate cases of placental insufficiency between 27 and 33 weeks. High abnormality scores correlate with low birth weight, premature birth and histopathological findings. Retrospective application on a different cohort imaged at 1.5 T illustrates the ability for direct clinical translation. CONCLUSION The presented automatic pipeline facilitates a fast, robust and reliable prediction of placental maturation. It yields human-interpretable and verifiable intermediate results and quantifies uncertainties on the cohort-level and for individual predictions. The proposed machine-learning pipeline runs in close to real-time and, deployed in clinical settings, has the potential to become a cornerstone of diagnosis and intervention of placental insufficiency. APPLAUSE generalizes to an independent cohort imaged at 1.5 T, demonstrating robustness to different operational and clinical environments.
Collapse
Affiliation(s)
- Maximilian Pietsch
- Centre for Medical Engineering, King's College London, London, UK; Centre for the Developing Brain, King's College London, London, UK.
| | - Alison Ho
- Department of Women and Children's Health, King's College London, London, UK
| | - Alessia Bardanzellu
- Centre for Medical Engineering, King's College London, London, UK; Centre for the Developing Brain, King's College London, London, UK
| | - Aya Mutaz Ahmad Zeidan
- Centre for Medical Engineering, King's College London, London, UK; Centre for the Developing Brain, King's College London, London, UK
| | - Lucy C Chappell
- Department of Women and Children's Health, King's College London, London, UK
| | - Joseph V Hajnal
- Centre for Medical Engineering, King's College London, London, UK; Centre for the Developing Brain, King's College London, London, UK
| | - Mary Rutherford
- Centre for Medical Engineering, King's College London, London, UK; Centre for the Developing Brain, King's College London, London, UK
| | - Jana Hutter
- Centre for Medical Engineering, King's College London, London, UK; Centre for the Developing Brain, King's College London, London, UK
| |
Collapse
|
40
|
Miodownik S, Pikovsky O, Erez O, Kezerle Y, Lavon O, Rabinovich A. Unfolding the pathophysiology of congenital thrombotic thrombocytopenic purpura in pregnancy: lessons from a cluster of familial cases. Am J Obstet Gynecol 2021; 225:177.e1-177.e15. [PMID: 33621545 DOI: 10.1016/j.ajog.2021.02.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 02/08/2021] [Accepted: 02/17/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Thrombotic thrombocytopenic purpura (TTP), a rare, potentially life-threatening thrombotic microangiopathy, manifests either as congenital TTP or acquired forms. It is caused by the absence or severe depletion of a disintegrin and metalloproteinase with thrombospondin motifs 13 (ADAMTS13) protease, leading to the accumulation of ultra large von Willebrand factor multimers as well as extensive platelet adhesion and clumping, which can ultimately cause severe secondary end-organ damage. Pregnancy can provoke or exacerbate TTP, leading to maternal and fetal complications. OBJECTIVE In this report, we focused on pregnancy outcomes in a recently recognized cohort of congenital TTP patients of Bedouin Arab descent in southern Israel who were all homozygous for a novel c.3772delA variant of the ADAMTS13 gene, leading to the clinical manifestations of TTP largely during pregnancy. STUDY DESIGN All patients presented in this study belong to 2 closely related families of Arab Bedouin descent and were found to be homozygous for a novel ADAMTS13-c.3772delA variant. The cohort consisted of 19 females; 16 of them had congenital TTP and had been pregnant and were thus included. Patient data were collected from electronic medical records. RESULTS Of note, 13 women from our cohort, who delivered 14 fetuses (owing to 1 twin pregnancy), were diagnosed with congenital TTP following complicated pregnancies, which included recurrent pregnancy loss, stillbirth, early onset preeclampsia (both mild and severe), hemolysis, elevated liver enzymes and low platelet count syndrome, intrauterine growth restriction with abnormal Doppler flow, preterm premature rupture of membranes, and a total perinatal mortality rate of 30.7% (4/13). An additional 3 women, who were diagnosed owing to complications outside of pregnancy and at older ages, experienced TTP during their pregnancies, which occurred before diagnosis. Subsequent pregnancies were treated with fresh frozen plasma leading to a 100% fetal survival rate in the pregnancies that reached fetal viability. All placentas had lesions consistent with maternal vascular underperfusion. However, the severity and frequency of these lesions were lower in the 8 placentas from pregnancies treated with fresh frozen plasma. CONCLUSION This case series details a distinctive cohort of congenital TTP patients, all homozygous for the same, novel ADAMTS13 variant, who presented with clinical complications during pregnancy and maternal vascular lesions of underperfusion in the placenta. Our findings imply that the variant identified in the ADAMTS13 gene in our cohort may have a specific functional impact on the placenta, and that treatment with fresh frozen plasma during pregnancy ameliorates the course of the disease, leading to a milder phenotype or a normal pregnancy in the majority of cases.
Collapse
|
41
|
Vidaeff AC, Saade GR, Sibai BM. Preeclampsia: The Need for a Biological Definition and Diagnosis. Am J Perinatol 2021; 38:976-982. [PMID: 31986536 DOI: 10.1055/s-0039-1701023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The centuries-old approach to the prevention of eclampsia and its associated maternal morbidity and mortality is based on the recognition of the presence of premonitory signs and symptoms such as hypertension and proteinuria. The spectrum of preceding signs and symptoms came to be known as preeclampsia, which is debatably considered to be an early stage on a clinical continuum possibly leading to eclampsia. The premonitory signs and symptoms were then construed as diagnostic criteria for the poorly understood syndrome of preeclampsia, and this led to a perpetual debate that remains subject to wide disagreement and periodic updates. In this commentary, we will draw attention to the fact that the criteria for preeclampsia should be viewed from the prism of a screening test rather than as diagnostic of a condition in itself. Focusing research on developing better diagnostic and screening methods for what is clinically important, namely maternal and perinatal morbidity and mortality from hypertensive disorders of pregnancy, a long overdue upgrade from what was possible centuries ago, will ultimately lead to better management approaches to what really matters.
Collapse
Affiliation(s)
- Alex C Vidaeff
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas
| | - George R Saade
- Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, Texas
| | - Baha M Sibai
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, UT Health, Houston, Texas
| |
Collapse
|
42
|
Redline RW, Ravishankar S, Bagby CM, Saab ST, Zarei S. Four major patterns of placental injury: a stepwise guide for understanding and implementing the 2016 Amsterdam consensus. Mod Pathol 2021; 34:1074-1092. [PMID: 33558658 DOI: 10.1038/s41379-021-00747-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/09/2021] [Accepted: 01/09/2021] [Indexed: 01/05/2023]
Abstract
The Amsterdam classification system defines four major patterns of placental injury, maternal vascular malperfusion, fetal vascular malperfusion, acute chorioamnionitis, and villitis of unknown etiology, and lists the histologic findings that characterize each. However, there continues to be uncertainty regarding specific definitions, histologic mimics, grading and staging, and what combination of findings is required to diagnose each pattern of injury in a reproducible fashion. The purpose of this review is to clarify some of these issues by suggesting a stepwise approach to more fully realize the potential of this new classification system. In our view, the critical steps for correctly identifying and communicating each pattern of injury are (1) familiarity with the underlying pathophysiology and known clinical associations, (2) incorporation of important gross findings, (3) learning to recognize underlying architectural alterations and defining features at low power, (4) using higher magnification to narrow the differential diagnosis and assess severity (grading) and duration (staging), and (5) adopting a template for generating standardized placental reports that succinctly provide useful information for patient care and research applications.
Collapse
Affiliation(s)
- Raymond W Redline
- Department of Pathology, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, USA. .,Department of Reproductive Biology, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| | - Sanjita Ravishankar
- Department of Pathology, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Christina M Bagby
- Department of Pathology, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Shahrazad T Saab
- Department of Pathology, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Shabnam Zarei
- Department of Pathology, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| |
Collapse
|
43
|
Yong HEJ, Chan SY. Current approaches and developments in transcript profiling of the human placenta. Hum Reprod Update 2021; 26:799-840. [PMID: 33043357 PMCID: PMC7600289 DOI: 10.1093/humupd/dmaa028] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 06/05/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The placenta is the active interface between mother and foetus, bearing the molecular marks of rapid development and exposures in utero. The placenta is routinely discarded at delivery, providing a valuable resource to explore maternal-offspring health and disease in pregnancy. Genome-wide profiling of the human placental transcriptome provides an unbiased approach to study normal maternal–placental–foetal physiology and pathologies. OBJECTIVE AND RATIONALE To date, many studies have examined the human placental transcriptome, but often within a narrow focus. This review aims to provide a comprehensive overview of human placental transcriptome studies, encompassing those from the cellular to tissue levels and contextualize current findings from a broader perspective. We have consolidated studies into overarching themes, summarized key research findings and addressed important considerations in study design, as a means to promote wider data sharing and support larger meta-analysis of already available data and greater collaboration between researchers in order to fully capitalize on the potential of transcript profiling in future studies. SEARCH METHODS The PubMed database, National Center for Biotechnology Information and European Bioinformatics Institute dataset repositories were searched, to identify all relevant human studies using ‘placenta’, ‘decidua’, ‘trophoblast’, ‘transcriptome’, ‘microarray’ and ‘RNA sequencing’ as search terms until May 2019. Additional studies were found from bibliographies of identified studies. OUTCOMES The 179 identified studies were classifiable into four broad themes: healthy placental development, pregnancy complications, exposures during pregnancy and in vitro placental cultures. The median sample size was 13 (interquartile range 8–29). Transcriptome studies prior to 2015 were predominantly performed using microarrays, while RNA sequencing became the preferred choice in more recent studies. Development of fluidics technology, combined with RNA sequencing, has enabled transcript profiles to be generated of single cells throughout pregnancy, in contrast to previous studies relying on isolated cells. There are several key study aspects, such as sample selection criteria, sample processing and data analysis methods that may represent pitfalls and limitations, which need to be carefully considered as they influence interpretation of findings and conclusions. Furthermore, several areas of growing importance, such as maternal mental health and maternal obesity are understudied and the profiling of placentas from these conditions should be prioritized. WIDER IMPLICATIONS Integrative analysis of placental transcriptomics with other ‘omics’ (methylome, proteome and metabolome) and linkage with future outcomes from longitudinal studies is crucial in enhancing knowledge of healthy placental development and function, and in enabling the underlying causal mechanisms of pregnancy complications to be identified. Such understanding could help in predicting risk of future adversity and in designing interventions that can improve the health outcomes of both mothers and their offspring. Wider collaboration and sharing of placental transcriptome data, overcoming the challenges in obtaining sufficient numbers of quality samples with well-defined clinical characteristics, and dedication of resources to understudied areas of pregnancy will undoubtedly help drive the field forward.
Collapse
Affiliation(s)
- Hannah E J Yong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
| | - Shiao-Yng Chan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore.,Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
44
|
Roell KR, Harmon QE, Klungsøyr K, Bauer AE, Magnus P, Engel SM. Clustering Longitudinal Blood Pressure Trajectories to Examine Heterogeneity in Outcomes Among Preeclampsia Cases and Controls. Hypertension 2021; 77:2034-2044. [PMID: 33813841 DOI: 10.1161/hypertensionaha.120.16239] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Kyle R Roell
- Department of Epidemiology (K.R.R., S.M.E.), University of North Carolina at Chapel Hill
| | - Quaker E Harmon
- Epidemiology Branch, National Institute of Environmental Health Sciences, Durham, NC (Q.E.H.)
| | - Kari Klungsøyr
- Department of Global Public Health and Primary Care, University of Bergen, Norway (K.K.).,Division for Mental and Physical Health, Norwegian Institute of Public Health, Bergen, Norway (K.K.)
| | - Anna E Bauer
- Perinatal Psychiatry Program, Department of Psychiatry (A.E.B.), University of North Carolina at Chapel Hill
| | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway (P.M.)
| | - Stephanie M Engel
- Department of Epidemiology (K.R.R., S.M.E.), University of North Carolina at Chapel Hill
| |
Collapse
|
45
|
Roberts JM, Rich-Edwards JW, McElrath TF, Garmire L, Myatt L. Subtypes of Preeclampsia: Recognition and Determining Clinical Usefulness. Hypertension 2021; 77:1430-1441. [PMID: 33775113 DOI: 10.1161/hypertensionaha.120.14781] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The concept that preeclampsia is a multisystemic syndrome is appreciated in both research and clinical care. Our understanding of pathophysiology recognizes the role of inflammation, oxidative and endoplasm reticulum stress, and angiogenic dysfunction. Yet, we have not progressed greatly toward clinically useful prediction nor had substantial success in prevention or treatment. One possibility is that the maternal syndrome may be reached through different pathophysiological pathways, that is, subtypes of preeclampsia, that in their specificity yield more clinical utility. For example, early and late onset preeclampsia are increasingly acknowledged as different pathophysiological processes leading to a common presentation. Other subtypes of preeclampsia are supported by disparate clinical outcomes, long-range prognosis, organ systems involved, and risk factors. These insights have been supplemented by discovery-driven methods, which cluster preeclampsia cases into groups indicating different pathophysiologies. In this presentation, we review likely subtypes based on current knowledge and suggest others. We present a consideration of the requirements for a clinically meaningful preeclampsia subtype. A useful subtype should (1) identify a specific pathophysiological pathway or (2) specifically indicate maternal or fetal outcome, (3) be recognizable in a clinically useful time frame, and (4) these results should be reproducible and generalizable (but at varying frequency) including in low resource settings. We recommend that the default consideration be that preeclampsia includes several subtypes rather than trying to force all cases into a single pathophysiological pathway. The recognition of subtypes and deciphering their different pathophysiologies will provide specific targets for prevention, prediction, and treatment directing personalized care.
Collapse
Affiliation(s)
- James M Roberts
- Magee-Womens Research Institute, Department of Obstetrics Gynecology and Reproductive Sciences, Epidemiology and Clinical and Translational Research, University of Pittsburgh (J.M.R.)
| | - Janet W Rich-Edwards
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA (J.W.R.-E.).,Division of Women's Health, Department of Medicine (J.W.R.-E.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Thomas F McElrath
- Division of Maternal-Fetal Medicine (T.F.M.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Lana Garmire
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan (L.G.)
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Moore Institute of Nutrition and Wellness, Oregon Health and Science University (L.M.)
| | | |
Collapse
|
46
|
Dancey S, Mery E, Esteves A, Oltean I, Hayawi L, Tang K, Bainbridge S, El Demellawy D. Placenta pathology in recipient versus donor oocyte derivation for in vitro fertilization in a setting of hypertensive disorders of pregnancy and IUGR. Placenta 2021; 108:114-121. [PMID: 33865182 DOI: 10.1016/j.placenta.2021.03.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Assisted reproductive technology including in vitro fertilization (IVF) and oocyte donation (OD) may increase risk for placenta-mediated diseases. Comprehensive analysis of histopathological placental lesions according to source of oocytes used in the IVF procedure - recipient derived (RD-IVF) vs oocyte donation (OD-IVF), has not been conducted in a population with a hypertensive disorder of pregnancy (HDP) and/or intrauterine growth restriction (IUGR). METHODS A retrospective cohort study of archived placenta specimens from RD-IVF and OD-IVF pregnancies affected by HDP and/or IUGR was conducted with blinded histopathological placental examination. Three categories of lesions were differentiated and defined as main outcomes: maternal vascular malperfusion (MVM), chronic inflammation, and fetal vascular malperfusion (FVM). To determine the relationship between conception method and placental lesions, multivariable regressions were performed with maternal age, gestational age, HDP, birth and placental weight percentiles as model covariates. RESULTS 115 placentas were included 83 (72.2%) RD-IVF, 32 (27.8%) OD-IVF. Adjusted OR (aOR) for conception method was 5.05 (95%CI 0.58-43.90, p=0.142) for MVM, 1.87 (95%CI 0.68-5.15, p=0.228) for chronic inflammatory and 0.61 (95%CI 0.15-2.37, p=0.471) for FVM lesions. Multiple gestation demonstrated borderline association with MVM (aOR=0.24, 95%CI 0.04-1.51, p=0.129) and total pathology score (aRR=0.79, 95%CI 0.62-1.01, p=0.058). Subgroup analysis suggested greater odds of villitis of unknown etiology (VUE) for OD-IVF (aOR=2.98, 95%CI 1.12-7.93, p=0.029). DISCUSSION Source of oocyte derivation demonstrated no evidence of association with main outcomes in cases of HDP and/or IUGR. Subgroup analysis demonstrated increased rates of inflammatory lesions for OD-IVF. Multiple gestation may be associated with decreased MVM and total lesions.
Collapse
Affiliation(s)
- Sonia Dancey
- Faculty of Medicine, University of Ottawa, 451 Smyth Rd, Ottawa, ON K1H 8M5, Canada.
| | - Erika Mery
- Faculty of Health Sciences, University of Ottawa, 25 University Private, Ottawa, ON K1N 7K4, Canada.
| | - Ashley Esteves
- Faculty of Medicine, University of Ottawa, 451 Smyth Rd, Ottawa, ON K1H 8M5, Canada.
| | - Irina Oltean
- Department of Pathology, University of Ottawa, Children's Hospital of Eastern Ontario, 401 Smyth Rd, Ottawa, ON K1H 8L1, Canada.
| | - Lamia Hayawi
- Clinical Research Unit, Children's Hospital of Eastern Ontario, 401 Smyth Rd, Ottawa, ON K1H 8L1, Canada.
| | - Ken Tang
- Clinical Research Unit, Children's Hospital of Eastern Ontario, 401 Smyth Rd, Ottawa, ON K1H 8L1, Canada.
| | - Shannon Bainbridge
- Faculty of Health Sciences, University of Ottawa, 25 University Private, Ottawa, ON K1N 7K4, Canada.
| | - Dina El Demellawy
- Department of Pathology, University of Ottawa, Children's Hospital of Eastern Ontario, 401 Smyth Rd, Ottawa, ON K1H 8L1, Canada.
| |
Collapse
|
47
|
Pro- and Anti-Angiogenic Markers as Clinical Tools for Suspected Preeclampsia with and without FGR near Delivery—A Secondary Analysis. REPRODUCTIVE MEDICINE 2021. [DOI: 10.3390/reprodmed2010003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Objective—the objective of this study was to assess the accuracy of placental growth factor (PlGF), soluble Fms-like Tyrosine Kinase 1 (sFlt-1), and endoglin (sEng) in the diagnosis of suspected preeclampsia (PE) with and without fetal growth restriction (FGR) near delivery. Methods—this is a secondary analysis of a dataset of 125 pregnant women presenting at the high risk pregnancy clinic with suspected PE, FGR or PE + FGR in the University Medical Center of Slovenia. The dataset included 31 PE cases, 16 FGR cases, 42 PE + FGR cases, 15 cases who developed with unrelated complications before 37 weeks (wks) (PTD), and 21 unaffected controls who delivered a healthy baby at term. We also analyzed a sub-group of women who delivered early (<34 wks) including 10 PE, 12 FGR, 28 PE + FGR, and six PTD. Clinical management adhered to hospital guidelines. Marker levels were extracted from the dataset and were used to develop Receiver Operating Characteristic (ROC) curves and to calculate the area under the curve (AUC), the detection rates (DRs), and the false positive rates (FPRs). Previously published marker cutoffs for yes/no admission to hospital wards were extracted from the literature. Negative and positive predictive values (NPVs and PPVs) were evaluated for their value in determining whether hospital admission was required. Non-parametric tests were applied for statistical analysis; p < 0.05 was considered significant. Results—near delivery, all the pro-and anti-angiogenic markers provided diagnostic (ROC = 1.00) accuracy for the early (<34 wks) group of FGR. Diagnostic or near diagnostic (ROC = 0.95) accuracy was achieved by all marker for early PE + FGR but lower accuracy was achieved for early PE. For all cases, all markers, especially PlGF reached diagnostic or near diagnostic accuracy for FGR and PE + FGR. At this accuracy level, they can contribute to the clinical management of FGR, and PE + FGR. All the markers were less accurate for all PE cases. The use of published cutoffs was adequate for clinical management of FGR, whether early or for all cases, using an NPV > 90%. For PE + FGR, the PPV value approached 100%, especially for early cases, and can thus be implemented in clinical management. Neither NPV nor PPV were high enough for managing all cases of PE. There was no added value in measuring the PlGF/(sFlt-1 + sEng) ratio. Conclusion—This is the first study on a Slovenian population. It shows that near-delivery angiogenic biomarkers tests may be useful for confirming the diseases in cases where there is a diagnostic doubt. However, the clinical use of the biomarkers needs to be weighed against resources available and degree of certainty of the diagnosis made with and without them for managing suspected FGR and PE + FGR requiring delivery <34 wks, where they are very accurate, and furthermore in the management of all cases of FGR and FGR+PE. The markers were less accurate for the clinical diagnosis of PE.
Collapse
|
48
|
Johnsen GM, Fjeldstad HES, Drabbels JJM, Haasnoot GW, Eikmans M, Størvold GL, Alnaes-Katjavivi P, Jacobsen DP, Scherjon SA, Redman CWG, Claas FHJ, Staff AC. A possible role for HLA-G in development of uteroplacental acute atherosis in preeclampsia. J Reprod Immunol 2021; 144:103284. [PMID: 33578175 DOI: 10.1016/j.jri.2021.103284] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 12/09/2020] [Accepted: 01/26/2021] [Indexed: 12/18/2022]
Abstract
HLA-G, a non-classical HLA molecule expressed by extravillous trophoblasts, plays a role in the maternal immune tolerance towards fetal cells. HLA-G expression is regulated by genetic polymorphisms in the 3' untranslated region (3'UTR). Low levels of HLA-G in the maternal circulation and placental tissue are linked to preeclampsia. Our objective was to investigate whether variants of the 3'UTR of the HLA-G gene in mother and fetus are associated with acute atherosis, a pregnancy specific arterial lesion of the decidua basalis that is prevalent in preeclampsia. Paired maternal and fetal DNA samples from 83 normotensive and 83 preeclamptic pregnancies were analyzed. We sequenced the part of the HLA-G 3'UTR containing a 14-bp insertion/deletion region and seven single nucleotide polymorphisms (SNPs). Associations with acute atherosis were tested by logistic regression. The frequency of heterozygosity for the 14-bp polymorphism (Ins/Del) and the +3142 SNP (C/G) variant in the fetus are associated with acute atherosis in preeclampsia (66.7 % vs. 39.6 %, p = 0.039, and 69.0 % vs. 43.4 %, p = 0.024). Furthermore, the fetal UTR-3 haplotype, which encompasses the 14-bp deletion and the +3142G variant, is associated with acute atherosis in preeclampsia (15 % vs. 3.8 %, p = 0.016). In conclusion, HLA-G polymorphisms in the fetus are associated with acute atherosis. We hypothesize that these polymorphisms lead to altered HLA-G expression in the decidua basalis, affecting local feto-maternal immune tolerance and development of acute atherosis.
Collapse
Affiliation(s)
- Guro M Johnsen
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway; Division of Obstetrics and Gyneacology, Oslo University Hospital, Norway; Institute for Experimental Medical Research, Oslo University Hospital, Norway.
| | - Heidi E S Fjeldstad
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway; Division of Obstetrics and Gyneacology, Oslo University Hospital, Norway
| | - Jos J M Drabbels
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Netherlands
| | - Geert W Haasnoot
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Netherlands
| | - Michael Eikmans
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Netherlands
| | - Gro L Størvold
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway; Division of Obstetrics and Gyneacology, Oslo University Hospital, Norway; Institute for Experimental Medical Research, Oslo University Hospital, Norway
| | | | - Daniel P Jacobsen
- Division of Obstetrics and Gyneacology, Oslo University Hospital, Norway
| | - Sicco A Scherjon
- Department of Obstetrics and Gynecology, University of Groningen, Netherlands
| | | | - Frans H J Claas
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Netherlands
| | - Anne Cathrine Staff
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway; Division of Obstetrics and Gyneacology, Oslo University Hospital, Norway
| |
Collapse
|
49
|
Barr LC, Pudwell J, Smith GN. Postpartum microvascular functional alterations following severe preeclampsia. Am J Physiol Heart Circ Physiol 2021; 320:H1393-H1402. [PMID: 33481699 DOI: 10.1152/ajpheart.00767.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Preeclampsia is associated with adverse maternal health outcomes later in life. Vascular endothelial dysfunction has been previously described following preeclampsia. We hypothesized that microvascular endothelial dysfunction associated with preeclampsia persists postpartum and may identify those at greatest risk of future cardiovascular disease. The objective of this study was to examine postpartum microvascular endothelial function in women after a pregnancy complicated by preeclampsia. Women with previous preeclampsia (n = 30) and normotensive controls (n = 30) between 6 mo and 5 yr postpartum were recruited. Severity of preeclampsia [severe (n = 16) and mild (n = 14)] was determined by standardized chart review. Microvascular reactivity in the forearm was measured with laser speckle contrast imaging, coupled with iontophoresis; endothelium-dependent and endothelium-independent vasodilation was induced with 1% acetylcholine and sodium nitroprusside solutions, respectively. A postocclusive reactive hyperemia test assessed vasodilatory response following three minutes of suprasystolic (200 mmHg) occlusion with a mechanized cuff. Women with prior severe preeclampsia exhibited significantly higher vasodilation to acetylcholine and sodium nitroprusside compared to controls (P < 0.01; P = 0.03) and prior mild preeclampsia (P = 0.03; P < 0.01). Neither the degree of postocclusive reactive hyperemia (P = 0.98), nor time to return halfway to baseline [OR = 1.026 (0.612, 1.72); P = 0.92], differed between preeclampsia and controls. In conclusion, severe preeclampsia is associated with heightened postpartum microvascular endothelium-dependent and endothelium-independent vasoreactivity. These changes, or a common antecedent, may be linked to postpartum alterations in vascular function that predispose women to disease after preeclampsia. Further investigation should identify the contributing mechanism and the degree to which it could be amenable to medical intervention.NEW & NOTEWORTHY We examine maternal microvascular function after preeclampsia, identifying heightened endothelium-dependent and endothelium-independent microvascular reactivity following severe disease. Our study represents a noteworthy addition to the existing literature with the use of a novel imaging modality, vascular perturbation, postpartum time point, and patient population with differentiation of preeclampsia into severe and nonsevere subtypes. These results represent a novel addition to the growing clinical and academic understanding of maternal health outcomes following preeclampsia.
Collapse
Affiliation(s)
- Logan C Barr
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jessica Pudwell
- Department of Obstetrics & Gynaecology, Queen's University, Kingston, Ontario, Canada
| | - Graeme N Smith
- Department of Obstetrics & Gynaecology, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
50
|
Li W, Chung CYL, Wang CC, Chan TF, Leung MBW, Chan OK, Wu L, Appiah K, Chaemsaithong P, Cheng YKY, Poon LCY, Leung TY. Monochorionic twins with selective fetal growth restriction: insight from placental whole-transcriptome analysis. Am J Obstet Gynecol 2020; 223:749.e1-749.e16. [PMID: 32437666 DOI: 10.1016/j.ajog.2020.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/24/2020] [Accepted: 05/05/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND The underlying pathomechanism in placenta-related selective fetal growth restriction in monochorionic diamniotic twin pregnancy is not known. OBJECTIVE This study aimed to investigate any differences in placental transcriptomic profile between the selectively growth-restricted twins and the normally grown cotwins in monochorionic diamniotic twin pregnancies. STUDY DESIGN This was a prospective study of monochorionic diamniotic twin pregnancies complicated by selective fetal growth restriction. Placental biopsy specimens were obtained from the subjects in the delivery suite. The placental transcriptome of the selectively growth-restricted twin was compared with that of the normally grown cotwin. This study was divided into 2 stages: (1) gene discovery phase in which placental tissues from 5 monochorionic diamniotic twin pregnancies complicated by selective fetal growth restriction plus 2 control twin pregnancies underwent transcriptome profiling, and transcriptome profiling was carried out using whole-genome RNA sequencing; and (2) validation phase in which placental tissues from 13 monochorionic diamniotic twin pregnancies with selective fetal growth restriction underwent RNA and protein validation. RNA and protein expression levels of candidate genes were determined using quantitative real-time polymerase chain reaction and immunohistochemistry staining. RESULTS A total of 1429 transcripts were differentially expressed in the placentae of selectively growth-restricted twin pairs, where 610 were up-regulated and 819 were down-regulated. Endoplasmic reticulum lectin and mannose 6-phosphate receptor were consistently differentially up-regulated in all placentae of selectively growth-restricted twins. Quantitative real-time polymerase chain reaction and immunohistochemistry staining were used to validate the results (P<.05). CONCLUSION The expression of endoplasmic reticulum lectin and mannose 6-phosphate receptor, which are important for angiogenesis and fetal growth, was significantly increased in the placentae of selectively growth-restricted twin of a monochorionic twin pair.
Collapse
Affiliation(s)
- Wei Li
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Claire Yik Lok Chung
- School of Life Sciences, the Chinese University of Hong Kong, Shatin, Hong Kong; Hong Kong Bioinformatics Centre, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong; Department of Reproduction and Development, Li Ka Shing Institute of Health Sciences, the Chinese University of Hong Kong, Shatin, Hong Kong; School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ting Fung Chan
- School of Life Sciences, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Maran Bo Wah Leung
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Oi Ka Chan
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong; Hong Kong Bioinformatics Centre, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ling Wu
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kubi Appiah
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Piya Chaemsaithong
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yvonne Kwun Yue Cheng
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Liona Chiu Yee Poon
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Tak Yeung Leung
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong.
| |
Collapse
|