1
|
Fierro JJ, Schoots MH, Liefers SC, der Meer BDV, Diercks GFH, Bootsma H, Prins JR, Westra J, de Leeuw K. Immunohistochemical analysis reveals higher Myxovirus resistance protein 1 expression and increased macrophage count in placentas from patients with systemic rheumatic diseases. Rheumatol Int 2025; 45:90. [PMID: 40183958 PMCID: PMC11971135 DOI: 10.1007/s00296-025-05856-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
To compare immune cell subsets and interferon (IFN) expression in placentas from patients with systemic lupus erythematosus (SLE), primary Sjögren's disease (pSjD), antiphospholipid syndrome (APS), healthy controls (HC) and of women with adverse pregnancy outcomes (APO) without these systemic rheumatic diseases (SRD). Placenta biopsies from HC, SLE, pSjD, APS, and patients with fetal growth restriction (FGR), spontaneous preterm birth (PTB), or FGR and preeclampsia (FGR/PE) attended between 2008 and 2022 were recovered from the pathology biobank of the University Medical Center Groningen. Clinical characteristics and APO were retrieved from medical records. Immunohistochemistry was performed for Myxovirus resistance protein 1 (MxA), CD3, CD20, CD56, CD68, CD123, and Foxp3. The proportion of positive cells was established using an automated detection classifier, while MxA expression was assessed semi/quantitatively discriminating between maternal (decidua) and fetal (villi) tissue. Finally, placental lesion classification was performed. Our study included placentas from 11 SLE, 4 pSjD, 8 APS, 4 PTB, 8 FGR, 8 FGR/PE patients and 11 HC. A high rate of APO (70%) was identified in SRD patients. Patients with SRD had a higher macrophage (CD68+) count in decidua and villi than HC, but no differences were observed in T (CD3+), B (CD20+), NK (CD56+) and T regulatory (Foxp3+) cell count. No plasmacytoid dendritic cells (CD123+) were identified. Furthermore, patients with these SRD had higher MxA values than HC in villi but not in decidua. SLE, pSjD and APS patients have an increased macrophage count and interferon upregulation in the placenta compared to HC. Therefore, a pro-inflammatory environment might be key inducing placental dysfunction, which may lead to subsequent APO development.
Collapse
Affiliation(s)
- Juan J Fierro
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9700RB, The Netherlands.
- Grupo Reproducción, Departamento de Microbiología y Parasitología, Universidad de Antioquia UdeA, Medellín, Colombia.
| | - Mirthe H Schoots
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Silvia C Liefers
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9700RB, The Netherlands
| | - Berber Doornbos-van der Meer
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9700RB, The Netherlands
| | - Gilles F H Diercks
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hendrika Bootsma
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9700RB, The Netherlands
| | - Jelmer R Prins
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Johanna Westra
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9700RB, The Netherlands
| | - Karina de Leeuw
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9700RB, The Netherlands
| |
Collapse
|
2
|
Moss CG, Dilworth MR, Harris LK, Freeman S, Heazell AEP. Understanding a Potential Role for the NLRP3 Inflammasome in Placenta-Mediated Pregnancy Complications. Am J Reprod Immunol 2025; 93:e70077. [PMID: 40260875 PMCID: PMC12013246 DOI: 10.1111/aji.70077] [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: 12/18/2024] [Revised: 03/20/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025] Open
Abstract
Stillbirth affects approximately 2 million pregnancies annually and is closely linked to placental dysfunction, which may also present clinically as foetal growth restriction (FGR) or pre-eclampsia (PE). Placental dysfunction can arise from a range of insults, including the inflammatory conditions villitis of unknown aetiology (VUE) and chronic histiocytic intervillositis (CHI). Despite ample research regarding the pathophysiology of placental dysfunction, the literature surrounding placental inflammation is more limited, with no currently established treatments. In the absence of infection, placental inflammation is hypothesised to be stimulated by damage-associated molecular patterns (DAMPs), known as sterile inflammation. The NLRP3 inflammasome, a protein scaffold that unites within the cytosol of cells, is a proposed contributor. The NLRP3 inflammasome is dysregulated in numerous diseases and has shown evidence of activation through the sterile inflammatory pathway via DAMPs. Studies have demonstrated the upregulation of the NLRP3 inflammasome and its components in placentally-mediated pregnancy pathologies. However, the link between placental dysfunction seen in these disorders and the NLRP3 inflammasome is not yet firmly established. This manuscript aims to review the evidence regarding placental inflammation seen with placental dysfunction, discuss its association with the NLRP3 inflammasome, and identify potential therapeutic interventions for this pathological inflammatory response.
Collapse
Affiliation(s)
- Chloe G. Moss
- Maternal and Fetal Health Research CentreDivision of Developmental Biology and MedicineUniversity of ManchesterManchesterUK
- Manchester Academic Health Science CentreManchester University NHS Foundation TrustManchesterUK
| | - Mark R. Dilworth
- Maternal and Fetal Health Research CentreDivision of Developmental Biology and MedicineUniversity of ManchesterManchesterUK
- Manchester Academic Health Science CentreManchester University NHS Foundation TrustManchesterUK
| | - Lynda K. Harris
- Department of Obstetrics and GynaecologyOlson Center for Women's HealthUniversity of Nebraska Medical CentreOmahaUSA
| | - Sally Freeman
- Division of Pharmacy and OptometryUniversity of ManchesterManchesterUK
| | - Alexander E. P. Heazell
- Maternal and Fetal Health Research CentreDivision of Developmental Biology and MedicineUniversity of ManchesterManchesterUK
- Manchester Academic Health Science CentreManchester University NHS Foundation TrustManchesterUK
| |
Collapse
|
3
|
Berns LL, Wiegel RE, Koning AH, Willemsen SP, Laven JS, Steegers-Theunissen RP. Utero-placental (vascular) development throughout pregnancy in women with polycystic ovary syndrome: the Rotterdam Periconceptional Cohort. F S Rep 2025; 6:79-89. [PMID: 40201084 PMCID: PMC11973822 DOI: 10.1016/j.xfre.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 04/10/2025] Open
Abstract
Objective To study utero-placental vascular development from the first trimester onward in pregnant women with polycystic ovary syndrome (PCOS) and successful live births, compared with pregnant women without PCOS, after in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) treatment or natural conception. Design Prospective periconceptional cohort study in a single tertiary hospital. Subjects Participants with ongoing pregnancies with available serial three-dimensional ultrasound scans were divided into 3 groups: women with PCOS; subfertile group, pregnancies who conceived via IVF/ICSI without PCOS; and fertile group, pregnancies who conceived naturally without PCOS. Exposure PCOS diagnosis. Main Outcome Measures During the first-trimester, placental volume (PV) and utero-placental vascular volume (uPVV) were measured offline in three-dimensional ultrasound volumes obtained at 7, 9, and 11 weeks' gestational age (GA) using Virtual Organ Analysis and Virtual Reality. Serial measurements were obtained from uterine artery pulsatility and resistance indices (UtA PI and UtA RI) measured by pulsed-wave Doppler ultrasound as well as mean arterial pressure at 7, 9, 11, 13, 22, and 32 weeks' GA. Similarly, the umbilical artery PI and RI were measured at 22 and 32 weeks' GA. Results We included 206 pregnancies in our study (PCOS n = 41; subfertile n = 63; fertile n = 102). Significantly negative associations were observed between PCOS and placental measurements (PV, uPVV, and their ratio) at 11 weeks' GA with both the subfertile and fertile group as reference (e.g., uPVV 11 weeks' GA: betaPCOS-fertile -0.18 ∛cm3 [95% confidence interval: -0.30; -0.06]). UtA PI and RI were significantly lower throughout pregnancy in women with PCOS compared with the subfertile and fertile group. Women with PCOS showed a negative association with umbilical artery PI and RI at 32 weeks' GA compared with the subfertile and fertile group as reference. Conclusion Women with PCOS show decreased first-trimester placental development at 11 weeks' GA compared with pregnancies without PCOS in the subfertile and fertile group. Additionally, these women also display lower UtA PI and UtA RI compared with women without PCOS. These results support the hypothesis that PCOS impacts early placental development, potentially contributing to adverse pregnancy outcomes. Further research should focus on the underlying pathophysiology and the modifying role of IVF/ICSI treatment.
Collapse
Affiliation(s)
- Lotte L. Berns
- Department of Obstetrics and Gynecology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Rosalieke E. Wiegel
- Department of Obstetrics and Gynecology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Anton H.J. Koning
- Department of Pathology, Clinical Bioinformatics Unit, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Sten P. Willemsen
- Department of Obstetrics and Gynecology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Biostatistics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Joop S.E. Laven
- Department of Obstetrics and Gynecology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | | |
Collapse
|
4
|
Jin H, Wang X, Li L, Rui C, Gan H, Wang Q, Tao F, Zhu Y. Integrated proteomic and transcriptomic landscape of human placenta in small for gestational age infants. iScience 2024; 27:111423. [PMID: 39687015 PMCID: PMC11648249 DOI: 10.1016/j.isci.2024.111423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 09/01/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Small for gestational age (SGA) infants affected by placental insufficiency are exposed to the risk of stillbirth and long-term complications. Based on RNA-seq and mass spectrometry, we identified dysregulated RNAs and proteins from the comparisons of SGA placental tissues and controls. We revealed two SGA-relevant co-expression modules (SRMs) that also significantly distinguished SGA from controls. Then we performed an integrated analysis of transcriptomic and proteomic profiles to trace their links to SGA as well as their significant correlations. For the core functional molecules we screened, we revealed their potential upstream regulators and validated them experimentally in an independent cohort. Overall, we pointed out insights into different molecular pathways for the pathological mechanisms of SGA and indicated potential target molecules that may be drivers of placental aberrations in the SGA infants.
Collapse
Affiliation(s)
- Heyue Jin
- Department of Maternal & Child and Adolescent Health, School of Public Health, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui 230032, China
- Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Xianyan Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Lingyu Li
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Chen Rui
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Hong Gan
- Department of Maternal & Child and Adolescent Health, School of Public Health, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui 230032, China
| | - Qunan Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Fangbiao Tao
- Department of Maternal & Child and Adolescent Health, School of Public Health, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui 230032, China
| | - Yumin Zhu
- Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| |
Collapse
|
5
|
Ulusoy CO, Kurt A, Seyhanli Z, Hizli B, Bucak M, Agaoglu RT, Oguz Y, Yucel KY. Role of Inflammatory Markers and Doppler Parameters in Late-Onset Fetal Growth Restriction: A Machine-Learning Approach. Am J Reprod Immunol 2024; 92:e70004. [PMID: 39422068 DOI: 10.1111/aji.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/21/2024] [Accepted: 10/05/2024] [Indexed: 10/19/2024] Open
Abstract
OBJECTIVES This study evaluates the association of novel inflammatory markers and Doppler parameters in late-onset FGR (fetal growth restriction), utilizing a machine-learning approach to enhance predictive accuracy. MATERIALS AND METHODS A retrospective case-control study was conducted at the Department of Perinatology, Ministry of Health Etlik City Hospital, Ankara, from 2023 to 2024. The study included 240 patients between 32 and 37 weeks of gestation, divided equally between patients diagnosed with late-onset FGR and a control group. We focused on novel inflammatory markers-systemic immune-inflammation index (SII), systemic inflammatory response index (SIRI), and neutrophil-percentage-to-albumin ratio (NPAR)-and their correlation with Doppler parameters of umbilical and uterine arteries. Machine-learning algorithms were employed to analyze the data collected, including demographic, neonatal, and clinical parameters, to develop a predictive model for FGR. RESULTS The machine-learning model, specifically the Random Forest algorithm, effectively integrated the inflammatory markers with Doppler parameters to predict FGR. NPAR showed a significant correlation with FGR presence, providing a robust tool in the predictive model (Accuracy 77%, area under the curve [AUC] 0.851). In contrast, SII and SIRI, while useful, did not achieve the same level of predictive accuracy (Accuracy 75% AUC 0.818 and Accuracy 73% AUC 0.793, respectively). The model highlighted the potential of combining ultrasound measurements with inflammatory markers to improve diagnostic accuracy for late-onset FGR. CONCLUSIONS This study illustrates the efficacy of integrating machines with traditional diagnostic methods to enhance the prediction of late-onset FGR. Further research with a larger cohort is recommended to validate these findings and refine the predictive model, which could lead to improved clinical outcomes for affected pregnancies. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT06372938.
Collapse
Affiliation(s)
- Can Ozan Ulusoy
- Department of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Ahmet Kurt
- Department of Obstetrics and Gynecology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Zeynep Seyhanli
- Department of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Burak Hizli
- Department of Obstetrics and Gynecology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Mevlut Bucak
- Department of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | | | - Yüksel Oguz
- Department of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | | |
Collapse
|
6
|
Bezemer RE, Faas MM, van Goor H, Gordijn SJ, Prins JR. Decidual macrophages and Hofbauer cells in fetal growth restriction. Front Immunol 2024; 15:1379537. [PMID: 39007150 PMCID: PMC11239338 DOI: 10.3389/fimmu.2024.1379537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/14/2024] [Indexed: 07/16/2024] Open
Abstract
Placental macrophages, which include maternal decidual macrophages and fetal Hofbauer cells, display a high degree of phenotypical and functional plasticity. This provides these macrophages with a key role in immunologically driven events in pregnancy like host defense, establishing and maintaining maternal-fetal tolerance. Moreover, placental macrophages have an important role in placental development, including implantation of the conceptus and remodeling of the intrauterine vasculature. To facilitate these processes, it is crucial that placental macrophages adapt accordingly to the needs of each phase of pregnancy. Dysregulated functionalities of placental macrophages are related to placental malfunctioning and have been associated with several adverse pregnancy outcomes. Although fetal growth restriction is specifically associated with placental insufficiency, knowledge on the role of macrophages in fetal growth restriction remains limited. This review provides an overview of the distinct functionalities of decidual macrophages and Hofbauer cells in each trimester of a healthy pregnancy and aims to elucidate the mechanisms by which placental macrophages could be involved in the pathogenesis of fetal growth restriction. Additionally, potential immune targeted therapies for fetal growth restriction are discussed.
Collapse
Affiliation(s)
- Romy Elisa Bezemer
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, Netherlands
| | - Marijke M Faas
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, Netherlands
| | - Sanne Jehanne Gordijn
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
| | - Jelmer R Prins
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
7
|
Andreescu M. Correlation Between Maternal-Fetus Interface and Placenta-Mediated Complications. Cureus 2024; 16:e62457. [PMID: 38882223 PMCID: PMC11180486 DOI: 10.7759/cureus.62457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2024] [Indexed: 06/18/2024] Open
Abstract
Pregnancy is a highly regulated biological phenomenon that involves the development of a semi-allogeneic fetus inside the uterus of the mother. The maternal-fetal interface is a critical junction where communication takes place between the fetal and maternal immune systems, which determine the outcome of the pregnancy. The interface is composed of the decidua and placenta. The main cells present at the maternal-fetal interface include invading trophoblasts, maternal immune cells, and decidual stromal cells. Although maternal tolerance is crucial for maintaining a successful pregnancy, the role of the placenta in pregnancy is also important. Dysregulation of the placenta leads to various placenta-mediated complications, such as preeclampsia, intrauterine growth restriction, and placental abruption. Although the exact mechanism involving these complications is unclear, research has elucidated various factors involved in these pregnancy disorders. This review aimed to provide a summary of the maternal-fetal interface and immune mechanisms involved in placenta-mediated complications.
Collapse
Affiliation(s)
- Mihaela Andreescu
- Faculty of Medicine, Titu Maiorescu University, Bucharest, ROU
- Hematology, Colentina Clinical Hospital, Bucharest, ROU
| |
Collapse
|
8
|
Hoffman MC, Hunter SJ, D'Alessandro A, Christians U, Law AJ, Freedman R. Maternal Plasma Choline during Gestation and Small for Gestational Age Infants. Am J Perinatol 2024; 41:e939-e948. [PMID: 36584689 PMCID: PMC11407527 DOI: 10.1055/s-0042-1759775] [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] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Small for gestational age (SGA) infants are at increased risk for neonatal morbidity and developmental problems in childhood. No current interventions during human pregnancy address this problem. This study investigated the possible relationship between maternal choline concentration during pregnancy and SGA infants. STUDY DESIGN Maternal plasma choline concentrations were sampled at 16 and 28 weeks' gestation from women in a public prenatal clinic. Additional factors assessed were maternal age, body mass index, infection, C-reactive protein, hair cortisol, and compliance with prenatal vitamins and folate. Infants below the 10th percentile for gestational age were classified as SGA. Binary logistic regression was used to identify significant associated factors in pregnancies resulting in SGA infants compared with pregnancies resulting in non-SGA infants. RESULTS Thirteen (8%) of 159 women had SGA infants. Maternal plasma choline concentrations were low for pregnant participants whose infants were SGA, with the 28-week concentration significantly lower compared with other participants. Plasma choline concentrations ≥7 μM at 28 weeks, consistent with a minimally adequate dietary intake of choline-containing foods, were achieved by only 2 (15%) of mothers with SGA infants, compared with 51% of mothers whose infants were not SGA. Choline concentrations <7 μM at 28 weeks' gestation were associated with an odds ratio for SGA of 16.6 (95% confidence interval: 1.5-189.2, p = 0.023). Other significant factors were female sex and maternal C-reactive protein plasma concentration during gestation. CONCLUSION This observational study suggests that higher maternal choline levels may influence the risk for SGA. Maternal plasma choline concentrations are not routinely available in clinical laboratories. However, plasma choline levels can be increased by the mothers' intake of choline or phosphatidylcholine supplements. No nutritional intervention is currently recommended to prevent SGA, but the evidence from this study suggests that further consideration of the role of maternal choline may be warranted. KEY POINTS · More females are small for gestational age.. · Low maternal choline is related to small infants.. · Maternal choline ≥7 μM at 28 weeks appears optimal..
Collapse
Affiliation(s)
- Maria C Hoffman
- Division of Maternal and Fetal Medicine, Departments of Obstetrics and Gynecology and Psychiatry, University of Colorado School of Medicine, Aurora, Colorado
| | - Sharon J Hunter
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, Colorado
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado
| | - Uwe Christians
- Department of Anesthesiology, iC42 Clinical Research and Development, University of Colorado School of Medicine, Aurora, Colorado
| | - Amanda J Law
- Department of Psychiatry, Cell and Developmental Biology, and Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Robert Freedman
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
9
|
Tang H, Li D, Peng J, Yang W, Zhang X, Li H. Potential Association of Gut Microbial Metabolism and Circulating mRNA Based on Multiomics Sequencing Analysis in Fetal Growth Restriction. Mediators Inflamm 2024; 2024:9986187. [PMID: 38716374 PMCID: PMC11074908 DOI: 10.1155/2024/9986187] [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] [Received: 12/06/2023] [Revised: 02/27/2024] [Accepted: 03/25/2024] [Indexed: 08/21/2024] Open
Abstract
Objective Fetal growth restriction (FGR) is a significant contributor to negative pregnancy and postnatal developmental outcomes. Currently, the exact pathological mechanism of FGR remains unknown. This study aims to utilize multiomics sequencing technology to investigate potential relationships among mRNA, gut microbiota, and metabolism in order to establish a theoretical foundation for diagnosing and understanding the molecular mechanisms underlying FGR. Methods In this study, 11 healthy pregnant women and nine pregnant women with FGR were divided into Control group and FGR group based on the health status. Umbilical cord blood, maternal serum, feces, and placental tissue samples were collected during delivery. RNA sequencing, 16S rRNA sequencing, and metabolomics methods were applied to analyze changes in umbilical cord blood circulating mRNA, fecal microbiota, and metabolites. RT-qPCR, ELISA, or western blot were used to detect the expression of top 5 differential circulating mRNA in neonatal cord blood, maternal serum, or placental tissue samples. Correlation between differential circulating mRNA, microbiota, and metabolites was analyzed by the Spearman coefficient. Results The top 5 mRNA genes in FGR were altered with the downregulation of TRIM34, DEFA3, DEFA1B, DEFA1, and QPC, and the upregulation of CHPT1, SMOX, FAM83A, GDF15, and NAPG in newborn umbilical cord blood, maternal serum, and placental tissue. The abundance of Bacteroides, Akkermansia, Eubacterium_coprostanoligenes_group, Phascolarctobacterium, Parasutterella, Odoribacter, Lachnospiraceae_UCG_010, and Dielma were significantly enriched in the FGR group. Metabolites such as aspartic acid, methionine, alanine, L-tryptophan, 3-methyl-2-oxovalerate, and ketoleucine showed notable functional alterations. Spearman correlation analysis indicated that metabolites like methionine and alanine, microbiota (Tyzzerella), and circulating mRNA (TRIM34, SMOX, FAM83A, NAPG) might play a role as mediators in the communication between the gut and circulatory system interaction in FGR. Conclusion Metabolites (METHIONINE, alanine) as well as microbiota (Tyzzerella) and circulating mRNA (TRIM34, SMOX, FAM83A, NAPG) were possible mediators that communicated the interaction between the gut and circulatory systems in FGR.
Collapse
Affiliation(s)
- Hui Tang
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Dan Li
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Jing Peng
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Weitao Yang
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Xian Zhang
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Hanmei Li
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| |
Collapse
|
10
|
Yadav D, Ostrea EM, Cheng CT, Kisseih E, Maddipati KR, Thomas RL. Effect of docosahexaenoic acid and olive oil supplementation on pup weight in alcohol-exposed pregnant rats. Front Pediatr 2024; 12:1334285. [PMID: 38638591 PMCID: PMC11024321 DOI: 10.3389/fped.2024.1334285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/20/2024] [Indexed: 04/20/2024] Open
Abstract
Background Low birth weight has been observed in offspring of alcoholic mothers due likely to unresolved inflammation and oxidative injury. Dietary lipids play a role in inflammation and its resolution. The primary objective was to investigate the effect of DHA and olive oil on the birth weight of pups born to alcohol-exposed dams. Methods Pregnant rats were randomized to the control or three treatment (alcohol) groups. From gestational days (GD) 8-19, the control group received daily olive oil and malto/dextrose, whereas groups 2 and 3 received olive oil and low-dose alcohol or high-dose alcohol, respectively. Group 4 received daily DHA and high-dose alcohol. The dam's blood was collected on GD 15 and 20 for cytokine analysis. Dams were sacrificed on GD 20. The mean birth weight of pups was compared by one-way ANOVA with post hoc Duncan's test. Results There was a significant increase in the pups' mean birth weight in the high-dose alcohol/DHA and high-dose alcohol/olive oil. Higher pro-inflammatory cytokines (IL-1β and IL-12p70) were noted in the alcohol-exposed dams. Conclusions DHA and olive oil supplementation in alcohol-exposed pregnant rats significantly increased their pups' birth weight despite having high pro-inflammatory cytokines. The mechanism of this effect remains to be determined.
Collapse
Affiliation(s)
- Deepak Yadav
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Enrique M. Ostrea
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Charlie T. Cheng
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Esther Kisseih
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Krishna R. Maddipati
- Bioactive Lipids Research Program, Wayne State University School of Medicine, Detroit, MI, United States
| | - Ronald L. Thomas
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
11
|
Bulka CM, Rajkotwala HM, Eaves LA, Gardner AJ, Parsons PJ, Galusha AL, O'Shea TM, Fry RC. Placental cellular composition and umbilical cord tissue metal(loid) concentrations: A descriptive molecular epidemiology study leveraging DNA methylation. Placenta 2024; 147:28-30. [PMID: 38281400 DOI: 10.1016/j.placenta.2024.01.009] [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: 09/20/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 01/30/2024]
Abstract
The placenta is a mixture of cell types, which may regulate maternal-fetal transfer of exogenous chemicals or become altered in response to exposures. We leveraged placental DNA methylation to characterize major constituent cell types and applied compositional data analysis to test associations with non-essential metal(loid)s measured in paired umbilical cord tissue (N = 158). Higher proportions of syncytiotrophoblasts were associated with lower arsenic, whereas higher proportions of Hofbauer cells were associated with higher cadmium concentrations in umbilical cords. These findings suggest that placental cellular composition influences amounts of metal(loid)s transferred to the fetus or that prenatal exposures alter the placental cellular makeup.
Collapse
Affiliation(s)
- Catherine M Bulka
- College of Public Health, University of South Florida, Tampa, FL, USA
| | | | - Lauren A Eaves
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Amaree J Gardner
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Patrick J Parsons
- Laboratory of Inorganic and Nuclear Chemistry, Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY, USA; Department of Environmental Health Sciences, School of Public Health, University at Albany, Rensselaer, NY, USA
| | - Aubrey L Galusha
- Laboratory of Inorganic and Nuclear Chemistry, Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY, USA; Department of Environmental Health Sciences, School of Public Health, University at Albany, Rensselaer, NY, USA
| | - T Michael O'Shea
- Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Rebecca C Fry
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA; Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
12
|
Yang J, Liu Y, Dong M. Integrated Bioinformatics Analysis to Screen Hub Gene Signatures for Fetal Growth Restriction. Genet Res (Camb) 2023; 2023:3367406. [PMID: 37033160 PMCID: PMC10079385 DOI: 10.1155/2023/3367406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/26/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023] Open
Abstract
Background. Fetal growth restriction (FGR) is the impairment of the biological growth potential of the fetus and often leads to adverse pregnancy outcomes. The molecular mechanisms for the development of FGR, however, are still unclear. The purpose of this study is to identify critical genes associated with FGR through an integrated bioinformatics approach and explore the potential pathogenesis of FGR. Methods. We downloaded FGR-related gene microarray data, used weighted gene co-expression network analysis (WGCNA), differentially expressed genes (DEGs), and protein-protein interaction (PPI) networks to screen hub genes. The GSE24129 gene set was used for validation of critical gene expression levels and diagnostic capabilities. Results. A weighted gene co-expression network was constructed, and 5000 genes were divided into 12 modules. Of these modules, the blue module showed the closest relationship with FGR. Taking the intersection of the DEGs and genes in the blue module as pivotal genes, 277 genes were identified, and 20 crucial genes were screened from the PPI network. The GSE24129 gene set verified the expression of 20 genes, and CXCL9, CXCR3, and ITGAX genes were identified as actual pivotal genes. The expression levels of CXCL9, CXCR3, and ITGAX were increased in both the training and validation sets, and ROC curve validation revealed that these three pivotal genes had a significant diagnostic ability for FGR. Single-gene GSEA results showed that all three core genes activated “hematopoietic cell lineage” and “cell adhesion molecules” and inhibited the “cGMP-PKG signaling pathway” in the development of FGR. CXCL9, CXCR3, and ITGAX may therefore be closely associated with the development of FGR and may serve as potential biomarkers for the diagnosis and treatment of FGR.
Collapse
|
13
|
Parcial ALN, Salomão NG, Portari EA, Arruda LV, de Carvalho JJ, de Matos Guedes HL, Conde TC, Moreira ME, Batista MM, Paes MV, Rabelo K, Gomes-Silva A. SARS-CoV-2 Is Persistent in Placenta and Causes Macroscopic, Histopathological, and Ultrastructural Changes. Viruses 2022; 14:1885. [PMID: 36146692 PMCID: PMC9500736 DOI: 10.3390/v14091885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/15/2022] [Accepted: 08/23/2022] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 is a virus that belongs to the Betacoronavirus genus of the Coronaviridae family. Other coronaviruses, such as SARS-CoV and MERS-CoV, were associated with complications in pregnant women. Therefore, this study aimed to report the clinical history of five pregnant women infected with SARS-CoV-2 (four symptomatic and one asymptomatic who gave birth to a stillborn child) during the COVID-19 pandemic. They gave birth between August 2020 to January 2021, a period in which there was still no vaccination for COVID-19 in Brazil. In addition, their placental alterations were later investigated, focusing on macroscopic, histopathological, and ultrastructural aspects compared to a prepandemic sample. Three of five placentas presented SARS-CoV-2 RNA detected by RT-PCRq at least two to twenty weeks after primary pregnancy infection symptoms, and SARS-CoV-2 spike protein was detected in all placentas by immunoperoxidase assay. The macroscopic evaluation of the placentas presented congested vascular trunks, massive deposition of fibrin, areas of infarctions, and calcifications. Histopathological analysis showed fibrin deposition, inflammatory infiltrate, necrosis, and blood vessel thrombosis. Ultrastructural aspects of the infected placentas showed a similar pattern of alterations between the samples, with predominant characteristics of apoptosis and detection of virus-like particles. These findings contribute to a better understanding of the consequences of SARS-CoV-2 infection in placental tissue, vertical transmission.
Collapse
Affiliation(s)
- André Luiz N. Parcial
- Interdisciplinary Laboratory of Medical Research, Instituto Oswaldo Cruz, Oswaldo Cruz Foundation, Rio de Janeiro 21040900, Brazil
| | - Natália Gedeão Salomão
- Interdisciplinary Laboratory of Medical Research, Instituto Oswaldo Cruz, Oswaldo Cruz Foundation, Rio de Janeiro 21040900, Brazil
| | - Elyzabeth Avvad Portari
- Pathological Anatomy, Fernandes Figueira Institute, Oswaldo Cruz Foundation, Rio de Janeiro 22250020, Brazil
| | - Laíza Vianna Arruda
- Interdisciplinary Laboratory of Medical Research, Instituto Oswaldo Cruz, Oswaldo Cruz Foundation, Rio de Janeiro 21040900, Brazil
| | - Jorge José de Carvalho
- Laboratory of Ultrastructure and Tissue Biology, Rio de Janeiro State University, Rio de Janeiro 20551030, Brazil
| | - Herbert Leonel de Matos Guedes
- Interdisciplinary Laboratory of Medical Research, Instituto Oswaldo Cruz, Oswaldo Cruz Foundation, Rio de Janeiro 21040900, Brazil
| | | | - Maria Elizabeth Moreira
- Pathological Anatomy, Fernandes Figueira Institute, Oswaldo Cruz Foundation, Rio de Janeiro 22250020, Brazil
| | - Marcelo Meuser Batista
- Pathological Anatomy, Fernandes Figueira Institute, Oswaldo Cruz Foundation, Rio de Janeiro 22250020, Brazil
| | - Marciano Viana Paes
- Interdisciplinary Laboratory of Medical Research, Instituto Oswaldo Cruz, Oswaldo Cruz Foundation, Rio de Janeiro 21040900, Brazil
| | - Kíssila Rabelo
- Laboratory of Ultrastructure and Tissue Biology, Rio de Janeiro State University, Rio de Janeiro 20551030, Brazil
| | - Adriano Gomes-Silva
- Interdisciplinary Laboratory of Medical Research, Instituto Oswaldo Cruz, Oswaldo Cruz Foundation, Rio de Janeiro 21040900, Brazil
- Mycobacteriosis Clinical Research Laboratory, National Institute of Infectious Diseases Evandro Chagas, Oswaldo Cruz Foundation, Rio de Janeiro 21040900, Brazil
| |
Collapse
|
14
|
Matulova J, Kacerovsky M, Hornychova H, Stranik J, Mls J, Spacek R, Burckova H, Jacobsson B, Musilova I. Acute Histological Chorioamnionitis and Birth Weight in Pregnancies With Preterm Prelabor Rupture of Membranes: A Retrospective Cohort Study. Front Pharmacol 2022; 13:861785. [PMID: 35308217 PMCID: PMC8931836 DOI: 10.3389/fphar.2022.861785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/14/2022] [Indexed: 11/18/2022] Open
Abstract
Aim: To assess the association between the birth weight of newborns from pregnancies with preterm prelabor rupture of membranes (PPROM) and the presence of acute histological chorioamnionitis (HCA) with respect to the: i) fetal and maternal inflammatory responses and ii) acute inflammation of the amnion. Material and Methods: This retrospective cohort study included 818 women with PPROM. A histopathological examination of the placenta was performed. Fetal inflammatory response was defined as the presence of any neutrophils in umbilical cord (histological grades 1–4) and/or chorionic vasculitis (histological grade 4 for the chorionic plate). Maternal inflammatory response was defined as the presence of histological grade 3–4 for the chorion-decidua and/or grade 3 for the chorionic plate and/or grade 1–4 for the amnion. Acute inflammation of the amnion was defined as the presence of any neutrophils in the amnion (histological grade 1–4 for the amnion). Birth weights of newborns were expressed as percentiles derived from INTERGROWTH-21st standards for the i) estimated fetal weight and ii) newborn birth weight. Results: No difference in percentiles of birth weights of newborns was found among the women with the women with HCA with fetal inflammatory response, with HCA with maternal inflammatory response and those without HCA. Women with HCA with acute inflammation of the amnion had lower percentiles of birth weights of newborns, derived from the estimated fetal weight standards, than women with HCA without acute inflammation of the amnion and those with the absence of HCA in the crude (with acute inflammation: median 46, without acute inflammation: median 52, the absence of HCA: median 55; p = 0.004) and adjusted (p = 0.02) analyses. The same subset of pregnancies exhibited the highest rate of newborns with a birth weight of ≤25 percentile. When percentiles were derived from the newborn weight standards, no differences in birth weights were observed among the subgroups. Conclusion: Acute inflammation of the amnion was associated with a lower birth weight in PPROM pregnancies, expressed as percentiles derived from the estimated fetal weight standards.
Collapse
Affiliation(s)
- Jana Matulova
- Department of Non-Medical Studies, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czechia
| | - Marian Kacerovsky
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czechia
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czechia
- *Correspondence: Marian Kacerovsky,
| | - Helena Hornychova
- Fingerland’s Institute of Pathology, University Hospital Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Jaroslav Stranik
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czechia
| | - Jan Mls
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czechia
| | - Richard Spacek
- Department of Obstetrics and Gynecology, University Hospital Ostrava, Ostrava, Czechia
| | - Hana Burckova
- Department of Neonatology, University Hospital Ostrava, Ostrava, Czechia
| | - Bo Jacobsson
- Department of Obstetrics and Gynecology, Institute of Clinical Science, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Obstetrics and Gynecology, Gothenburg, Sweden
- Department of Genetics and Bioinformatics, Domain of Health Data and Digitalization, Institute of Public Health, Oslo, Norway
| | - Ivana Musilova
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czechia
| |
Collapse
|
15
|
Broekhuizen M, Hitzerd E, van den Bosch TPP, Dumas J, Verdijk RM, van Rijn BB, Danser AHJ, van Eijck CHJ, Reiss IKM, Mustafa DAM. The Placental Innate Immune System Is Altered in Early-Onset Preeclampsia, but Not in Late-Onset Preeclampsia. Front Immunol 2022; 12:780043. [PMID: 34992598 PMCID: PMC8724430 DOI: 10.3389/fimmu.2021.780043] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/29/2021] [Indexed: 12/18/2022] Open
Abstract
Preeclampsia is a severe placenta-related pregnancy disorder that is generally divided into two subtypes named early-onset preeclampsia (onset <34 weeks of gestation), and late-onset preeclampsia (onset ≥34 weeks of gestation), with distinct pathophysiological origins. Both forms of preeclampsia have been associated with maternal systemic inflammation. However, alterations in the placental immune system have been less well characterized. Here, we studied immunological alterations in early- and late-onset preeclampsia placentas using a targeted expression profile approach. RNA was extracted from snap-frozen placenta samples (healthy n=13, early-onset preeclampsia n=13, and late-onset preeclampsia n=6). The expression of 730 immune-related genes from the Pan Cancer Immune Profiling Panel was measured, and the data were analyzed in the advanced analysis module of nSolver software (NanoString Technology). The results showed that early-onset preeclampsia placentas displayed reduced expression of complement, and toll-like receptor (TLR) associated genes, specifically TLR1 and TLR4. Mast cells and M2 macrophages were also decreased in early-onset preeclampsia compared to healthy placentas. The findings were confirmed by an immunohistochemistry approach using 20 healthy, 19 early-onset preeclampsia, and 10 late-onset preeclampsia placentas. We conclude that the placental innate immune system is altered in early-onset preeclampsia compared to uncomplicated pregnancies. The absence of these alterations in late-onset preeclampsia placentas indicates dissimilar immunological profiles. The study revealed distinct pathophysiological processes in early-onset and late-onset preeclampsia placentas and imply that a tailored treatment to each subtype is desirable.
Collapse
Affiliation(s)
- Michelle Broekhuizen
- Division of Neonatology, Department of Pediatrics, Erasmus University Medical Center, Rotterdam, Netherlands.,Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands.,Division of Experimental Cardiology, Department of Cardiology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Emilie Hitzerd
- Division of Neonatology, Department of Pediatrics, Erasmus University Medical Center, Rotterdam, Netherlands.,Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | - Jasper Dumas
- Department of Pathology, Erasmus University Medical Center, Rotterdam, Netherlands.,The Tumor Immuno-Pathology (TIP) Laboratory, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Robert M Verdijk
- Department of Pathology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Bas B van Rijn
- Department of Obstetrics and Gynecology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Casper H J van Eijck
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Irwin K M Reiss
- Division of Neonatology, Department of Pediatrics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Dana A M Mustafa
- Department of Pathology, Erasmus University Medical Center, Rotterdam, Netherlands.,The Tumor Immuno-Pathology (TIP) Laboratory, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
16
|
Brien ME, Gaudreault V, Hughes K, Hayes DJL, Heazell AEP, Girard S. A Systematic Review of the Safety of Blocking the IL-1 System in Human Pregnancy. J Clin Med 2021; 11:jcm11010225. [PMID: 35011965 PMCID: PMC8745599 DOI: 10.3390/jcm11010225] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/26/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
Blockade of the interleukin-1 (IL-1) pathway has been used therapeutically in several inflammatory diseases including arthritis and cryopyrin-associated periodic syndrome (CAPS). These conditions frequently affect women of childbearing age and continued usage of IL-1 specific treatments throughout pregnancy has been reported. IL-1 is involved in pregnancy complications and its blockade could have therapeutic potential. We systematically reviewed all reported cases of IL-1 blockade in human pregnancy to assess safety and perinatal outcomes. We searched several databases to find reports of specific blockade of the IL-1 pathway at any stage of pregnancy, excluding broad spectrum or non-specific anti-inflammatory intervention. Our literature search generated 2439 references of which 22 studies included, following extensive review. From these, 88 different pregnancies were assessed. Most (64.8%) resulted in healthy term deliveries without any obstetrical/neonatal complications. Including pregnancy exposed to Anakinra or Canakinumab, 12 (15.0%) resulted in preterm birth and one stillbirth occurred. Regarding neonatal complications, 2 cases of renal agenesis (2.5%) were observed, and 6 infants were diagnosed with CAPS (7.5%). In conclusion, this systematic review describes that IL-1 blockade during pregnancy is not associated with increased adverse perinatal outcomes, considering that treated women all presented an inflammatory disease associated with elevated risk of pregnancy complications.
Collapse
Affiliation(s)
- Marie-Eve Brien
- Ste-Justine Hospital Research Center, Montreal, QC H3T 1C5, Canada; (M.-E.B.); (V.G.); (K.H.)
| | - Virginie Gaudreault
- Ste-Justine Hospital Research Center, Montreal, QC H3T 1C5, Canada; (M.-E.B.); (V.G.); (K.H.)
| | - Katia Hughes
- Ste-Justine Hospital Research Center, Montreal, QC H3T 1C5, Canada; (M.-E.B.); (V.G.); (K.H.)
| | - Dexter J. L. Hayes
- Maternal and Fetal Health Research Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK; (D.J.L.H.); (A.E.P.H.)
| | - Alexander E. P. Heazell
- Maternal and Fetal Health Research Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK; (D.J.L.H.); (A.E.P.H.)
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Universite de Montreal, Montreal, QC H3T 1J4, Canada
- Department of Obstetrics and Gynecology, Department of Immunology, Mayo Clinic, Rochester, MN 55902, USA
- Correspondence: ; Tel.: +1-507-284-0545
| |
Collapse
|
17
|
Lean SC, Jones RL, Roberts SA, Heazell AEP. A prospective cohort study providing insights for markers of adverse pregnancy outcome in older mothers. BMC Pregnancy Childbirth 2021; 21:706. [PMID: 34670515 PMCID: PMC8527686 DOI: 10.1186/s12884-021-04178-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 09/28/2021] [Indexed: 11/26/2022] Open
Abstract
Background Advanced maternal age (≥35 years) is associated with increased rates of adverse pregnancy outcome. Better understanding of underlying pathophysiological processes may improve identification of older mothers who are at greatest risk. This study aimed to investigate changes in oxidative stress and inflammation in older women and identify clinical and biochemical predictors of adverse pregnancy outcome in older women. Methods The Manchester Advanced Maternal Age Study (MAMAS) was a multicentre, observational, prospective cohort study of 528 mothers. Participants were divided into three age groups for comparison 20–30 years (n = 154), 35–39 years (n = 222) and ≥ 40 years (n = 152). Demographic and medical data were collected along with maternal blood samples at 28 and 36 weeks’ gestation. Multivariable analysis was conducted to identify variables associated with adverse outcome, defined as one or more of: small for gestational age (< 10th centile), FGR (<5th centile), stillbirth, NICU admission, preterm birth < 37 weeks’ gestation or Apgar score < 7 at 5 min. Biomarkers of inflammation, oxidative stress and placental dysfunction were quantified in maternal serum. Univariate and multivariable logistic regression was used to identify associations with adverse fetal outcome. Results Maternal smoking was associated with adverse outcome irrespective of maternal age (Adjusted Odds Ratio (AOR) 4.22, 95% Confidence Interval (95%CI) 1.83, 9.75), whereas multiparity reduced the odds (AOR 0.54, 95% CI 0.33, 0.89). In uncomplicated pregnancies in older women, lower circulating anti-inflammatory IL-10, IL-RA and increased antioxidant capacity (TAC) were seen. In older mothers with adverse outcome, TAC and oxidative stress markers were increased and levels of maternal circulating placental hormones (hPL, PlGF and sFlt-1) were reduced (p < 0.05). However, these biomarkers only had modest predictive accuracy, with the largest area under the receiver operator characteristic (AUROC) of 0.74 for placental growth factor followed by TAC (AUROC = 0.69). Conclusions This study identified alterations in circulating inflammatory and oxidative stress markers in older women with adverse outcome providing preliminary evidence of mechanistic links. Further, larger studies are required to determine if these markers can be developed into a predictive model of an individual older woman’s risk of adverse pregnancy outcome, enabling a reduction in stillbirth rates whilst minimising unnecessary intervention. Supplementary Information The online version contains supplementary material available at 10.1186/s12884-021-04178-6.
Collapse
Affiliation(s)
- Samantha C Lean
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, St. Mary's Hospital, 5th Floor (Research), Oxford Road, Manchester, M13 9WL, UK
| | - Rebecca L Jones
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, St. Mary's Hospital, 5th Floor (Research), Oxford Road, Manchester, M13 9WL, UK
| | - Stephen A Roberts
- Centre for Biostatistics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Alexander E P Heazell
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, St. Mary's Hospital, 5th Floor (Research), Oxford Road, Manchester, M13 9WL, UK.
| |
Collapse
|
18
|
Matulova J, Kacerovsky M, Bolehovska R, Stranik J, Spacek R, Burckova H, Jacobsson B, Musilova I. Birth weight and intra-amniotic inflammatory and infection-related complications in pregnancies with preterm prelabor rupture of membranes: a retrospective cohort study. J Matern Fetal Neonatal Med 2021; 35:7571-7581. [PMID: 34320890 DOI: 10.1080/14767058.2021.1956458] [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] [Indexed: 01/02/2023]
Abstract
OBJECTIVE To assess the association between the birth weight of newborns and microbial invasion of the amniotic cavity (MIAC) and/or intra-amniotic inflammation in pregnancies with preterm prelabor rupture of membranes. METHODS A total of 528 pregnancies with preterm prelabor rupture of membranes were included in this retrospective cohort study. Transabdominal amniocentesis to determine the presence of MIAC (through culturing and molecular biology methods) and intra-amniotic inflammation (according to amniotic fluid interleukin-6 level) was performed as part of standard clinical management. Based on the presence of MIAC and/or intra-amniotic inflammation, the participants were divided into four subgroups: with intra-amniotic infection (presence of both), with sterile IAI (intra-amniotic inflammation alone), with colonization (MIAC alone), and with negative amniotic fluid (absence of both). Birth weights of newborns are expressed as percentiles derived from INTERGROWTH-21st standards for (i) newborn birth weight and (ii) estimated fetal weight. RESULTS No differences in birth weights, expressed as percentiles derived from newborn weight standards (infection: median 52; sterile: median 54; colonization: median 50; negative amniotic fluid: median 51; p = .93) and estimated fetal weight standards (infection: median 47; sterile: median 51; colonization: median 47; negative amniotic fluid: median 53; p = .48) were found among the four subgroups. No differences in percentiles (derived from both standards) were found in the subset of participants who delivered within 72 h after rupture of membranes (newborn weight standard, p = .99; estimated fetal weight standard, p = .81). CONCLUSIONS No association was identified between the birth weight of newborns and the presence of intra-amniotic inflammatory and infection-related complications in pregnancies with preterm prelabor rupture of membranes.
Collapse
Affiliation(s)
- Jana Matulova
- Department of Non-medical Studies, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czechia
| | - Marian Kacerovsky
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czechia.,Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Radka Bolehovska
- Institute of Clinical Biochemistry and Diagnostics, University Hospital Hradec Kralove, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czechia
| | - Jaroslav Stranik
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czechia
| | - Richard Spacek
- Department of Obstetrics and Gynecology, University Hospital Ostrava, Ostrava, Czechia
| | - Hana Burckova
- Department of Neonatology, University Hospital Ostrava, Ostrava, Czechia
| | - Bo Jacobsson
- Department of Obstetrics and Gynecology, Institute of Clinical Science, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Obstetrics and Gynecology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Genetics and Bioinformatics, Domain of Health Data and Digitalization, Institute of Public Health, Oslo, Norway
| | - Ivana Musilova
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czechia
| |
Collapse
|
19
|
Molecular Pathology Analysis of SARS-CoV-2 in Syncytiotrophoblast and Hofbauer Cells in Placenta from a Pregnant Woman and Fetus with COVID-19. Pathogens 2021; 10:pathogens10040479. [PMID: 33920814 PMCID: PMC8071113 DOI: 10.3390/pathogens10040479] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023] Open
Abstract
A small number of neonates delivered to women with SARS-CoV-2 infection have been found to become infected through intrauterine transplacental transmission. These cases are associated with a group of unusual placental pathology abnormalities that include chronic histiocytic intervillositis, syncytiotrophoblast necrosis, and positivity of the syncytiotrophoblast for SARS-CoV-2 antigen or RNA. Hofbauer cells constitute a heterogeneous group of immunologically active macrophages that have been involved in transplacental infections that include such viral agents as Zika virus and human immunodeficiency virus. The role of Hofbauer cells in placental infection with SARS-CoV-2 and maternal-fetal transmission is unknown. This study uses molecular pathology techniques to evaluate the placenta from a neonate infected with SARS-CoV-2 via the transplacental route to determine whether Hofbauer cells have evidence of infection. We found that the placenta had chronic histiocytic intervillositis and syncytiotrophoblast necrosis, with the syncytiotrophoblast demonstrating intense positive staining for SARS-CoV-2. Immunohistochemistry using the macrophage marker CD163, SARS-CoV-2 nucleocapsid protein, and double staining for SARS-CoV-2 with RNAscope and anti-CD163 antibody, revealed that no demonstrable virus could be identified within Hofbauer cells, despite these cells closely approaching the basement membrane zone of the infected trophoblast. Unlike some other viruses, there was no evidence from this transmitting placenta for infection of Hofbauer cells with SARS-CoV-2.
Collapse
|
20
|
Hypoxia and oxidative stress induce sterile placental inflammation in vitro. Sci Rep 2021; 11:7281. [PMID: 33790316 PMCID: PMC8012380 DOI: 10.1038/s41598-021-86268-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/08/2021] [Indexed: 12/18/2022] Open
Abstract
Fetal growth restriction (FGR) and stillbirth are associated with placental dysfunction and inflammation and hypoxia, oxidative and nitrative stress are implicated in placental damage. Damage-associated molecular patterns (DAMPs) are elevated in pregnancies at increased risk of FGR and stillbirth and are associated with increase in pro-inflammatory placental cytokines. We hypothesised that placental insults lead to release of DAMPs, promoting placental inflammation. Placental tissue from uncomplicated pregnancies was exposed in vitro to hypoxia, oxidative or nitrative stress. Tissue production and release of DAMPs and cytokines was determined. Oxidative stress and hypoxia caused differential release of DAMPs including uric acid, HMGB1, S100A8, cell-free fetal DNA, S100A12 and HSP70. After oxidative stress pro-inflammatory cytokines (IL-1α, IL-1β, IL-6, IL-8, TNFα, CCL2) were increased both within explants and in conditioned culture medium. Hypoxia increased tissue IL-1α/β, IL-6, IL-8 and TNFα levels, and release of IL-1α, IL-6 and IL-8, whereas CCL2 and IL-10 were reduced. IL1 receptor antagonist (IL1Ra) treatment prevented hypoxia- and oxidative stress-induced IL-6 and IL-8 release. These findings provide evidence that relevant stressors induce a sterile inflammatory profile in placental tissue which can be partially blocked by IL1Ra suggesting this agent has translational potential to prevent placental inflammation evident in FGR and stillbirth.
Collapse
|
21
|
Liao Z, Liu C, Cai L, Shen L, Sui C, Zhang H, Qian K. The Effect of Endometrial Thickness on Pregnancy, Maternal, and Perinatal Outcomes of Women in Fresh Cycles After IVF/ICSI: A Systematic Review and Meta-Analysis. Front Endocrinol (Lausanne) 2021; 12:814648. [PMID: 35222264 PMCID: PMC8874279 DOI: 10.3389/fendo.2021.814648] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 12/30/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Thin endometrium on ovulation triggering day is associated with impaired pregnancy outcomes in women after in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI), but the role of thick endometrium on pregnancy outcomes remains controversial. Moreover, there has been insufficient evidence currently to analyze the influence of endometrial thickness (EMT) on obstetric complications and perinatal outcomes. Thus, we performed this meta-analysis to evaluate the effect of EMT on pregnancy, maternal, and perinatal outcomes in an enlarged sample size. METHODS The databases Pubmed, Embase, Cochrane Libraries, and Web of Science were searched for English articles evaluating the correlation between EMT and pregnancy, maternal, or perinatal outcomes in women who underwent IVF/ICSI. We included studies that depicted a clear definition of outcomes and EMT grouping on ovulation triggering day. The EMT effect was analyzed in fresh cycle. Qualities of studies were assessed by the Newcastle-Ottawa Scale (NOS). Odds ratios (ORs) and weighted mean difference (WMD) with 95% confidence intervals (CIs) were calculated for analyzing dichotomous and continuous outcomes respectively, under a fixed or random effect model. RESULTS A total of 22 pieces of literature were included for the final meta-analysis. A decreased trend towards pregnancy outcomes was observed, such as live birth rate (LBR), clinical pregnancy rate (CPR), and implantation rate (IR) in the thin endometrium groups (EMT <7 mm). In contrast, thick endometrium (EMT >14 mm) had no effect on pregnancy outcomes compared to medium EMT groups (EMT 7-14 mm). Moreover, thin endometrium (EMT <7.5 mm) enhanced the incidence of hypertensive disorders of pregnancy (HDP) and small-for-gestational-age (SGA) infants, and decreased the birthweight (BW) of babies. CONCLUSIONS Our studies indicated that thin endometrium not only had detrimental effect on pregnancy outcomes, but also increased the risk of HDP in women and SGA of babies, or decreased BW of babies. The thick endometrium does not have an adverse effect on IVF outcomes. Therefore, patients need to be informed on possible obstetric complications and perinatal outcomes caused by thin endometrium and are encouraged to actively cooperate with perinatal care. SYSTEMATIC REVIEW REGISTRATION (https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=242637), identifier CRD42021242637.
Collapse
Affiliation(s)
- Zhiqi Liao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chang Liu
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical College, Nanjing, China
- *Correspondence: Chang Liu, ; Hanwang Zhang, ; Kun Qian,
| | - Lei Cai
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Shen
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong Sui
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hanwang Zhang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Chang Liu, ; Hanwang Zhang, ; Kun Qian,
| | - Kun Qian
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Chang Liu, ; Hanwang Zhang, ; Kun Qian,
| |
Collapse
|
22
|
Bezemer RE, Schoots MH, Timmer A, Scherjon SA, Erwich JJHM, van Goor H, Gordijn SJ, Prins JR. Altered Levels of Decidual Immune Cell Subsets in Fetal Growth Restriction, Stillbirth, and Placental Pathology. Front Immunol 2020; 11:1898. [PMID: 32973787 PMCID: PMC7468421 DOI: 10.3389/fimmu.2020.01898] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/15/2020] [Indexed: 12/14/2022] Open
Abstract
Immune cells are critically involved in placental development and functioning, and inadequate regulation of the maternal immune system is associated with placental pathology and pregnancy complications. This study aimed to explore numbers of decidual immune cells in pregnancies complicated with fetal growth restriction (FGR) and stillbirth (SB), and in placentas with histopathological lesions: maternal vascular malperfusion (MVM), fetal vascular malperfusion (FVM), delayed villous maturation (DVM), chorioamnionitis (CA), and villitis of unknown etiology (VUE). Placental tissue from FGR (n = 250), SB (n = 64), and healthy pregnancies (n = 42) was included. Histopathological lesions were classified according to criteria developed by the Amsterdam Placental Workshop Group. Tissue slides were stained for CD68 (macrophages), CD206 (M2-like macrophages), CD3 (T cells), FOXP3 [regulatory T (Treg) cells], and CD56 [natural killer (NK) cells]. Cell numbers were analyzed in the decidua basalis using computerized morphometry. The Mann-Whitney U-test and Kruskal Wallis test with the Dunn's as post-hoc test were used for statistical analysis. Numbers of CD68+ macrophages were higher in FGR compared to healthy pregnancies (p < 0.001), accompanied by lower CD206+/CD68+ ratios (p < 0.01). In addition, in FGR higher numbers of FOXP3+ Treg cells were seen (p < 0.01) with elevated FOXP3+/CD3+ ratios (p < 0.01). Similarly, in SB elevated FOXP3+ Treg cells were found (p < 0.05) with a higher FOXP3+/CD3+ ratio (p < 0.01). Furthermore, a trend toward higher numbers of CD68+ macrophages was found (p < 0.1) in SB. Numbers of CD3+ and FOXP3+ cells were higher in placentas with VUE compared to placentas without lesions (p < 0.01 and p < 0.001), accompanied by higher FOXP3+/CD3+ ratios (p < 0.01). Elevated numbers of macrophages with a lower M2/total macrophage ratio in FGR suggest a role for a macrophage surplus in its pathogenesis and could specifically indicate involvement of inflammatory macrophages. Higher numbers of FOXP3+ Treg cells with higher Treg/total T cell ratios in VUE may be associated with impaired maternal-fetal tolerance and a compensatory response of Treg cells. The abundant presence of placental lesions in the FGR and SB cohorts might explain the increase of Treg/total T cell ratios in these groups. More functionality studies of the observed altered immune cell subsets are needed.
Collapse
Affiliation(s)
- Romy E Bezemer
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Mirthe H Schoots
- Division of Pathology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Albertus Timmer
- Division of Pathology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Sicco A Scherjon
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jan Jaap H M Erwich
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Harry van Goor
- Division of Pathology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Sanne J Gordijn
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jelmer R Prins
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|