1
|
Wu YX, Li MJ, Liu Y, Guo M, Lan MN, Zheng HJ. ASPG and DAD1 are potential placental-derived biomarkers for ASD-like symptom severity levels in male/female offspring. Placenta 2024; 155:78-87. [PMID: 39154487 DOI: 10.1016/j.placenta.2024.08.009] [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: 05/09/2024] [Revised: 07/28/2024] [Accepted: 08/13/2024] [Indexed: 08/20/2024]
Abstract
INTRODUCTION An early evaluating system for autism spectrum disorder (ASD) severity is crucial. Questionnaire survey is challenging for accurately assessing the severity levels for ASD in children. METHODS Offspring with ASD-like phenotypes were induced by treating pregnant mice with Poly (I:C) at GD12.5 and the placentae corresponding to the offspring were obtained by caesarean. The autism severity composite score (ASCS) for offspring was calculated through behavioral tests. HE staining and immunohistochemistry were used to observe the morphology of placenta. Candidate biomarkers were identified by weighted protein co-expression network analysis (WPCNA) combined with machine learning and further validated by ELISA. Sperman's was used to analyze the correlation between biomarkers and metabolome. RESULTS The placental weight and mean vascular area of male offspring with ASD-like phenotypes were significantly decreased compared with typical mice. According to the WPCNA, four modules were identified and significantly correlated with ASCS of offspring. Two biomarkers (ASPG and DAD1) with high correlation with ASCS in offspring were identified. DISCUSSION VEGF pathway may contribute to sexual dimorphism in placental morphology within mice with ASD-like phenotypes in term. The placental ASPG and DAD1 levels could reflect ASD-like symptom severity levels in male/female mice offspring.
Collapse
Affiliation(s)
- Yi-Xiao Wu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Ming-Jie Li
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China; College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Yue Liu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Min Guo
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Meng-Ning Lan
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Hua-Jun Zheng
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
2
|
Li MR, Chen EX, Li ZH, Song HL, Zhang Y, Li FF, Xie YL, Tang J, Ding YB, Fu LJ. HMGB1 regulates autophagy of placental trophoblast through ERK signaling pathway†. Biol Reprod 2024; 111:414-426. [PMID: 38647664 DOI: 10.1093/biolre/ioae064] [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: 10/09/2023] [Revised: 12/21/2023] [Accepted: 04/18/2024] [Indexed: 04/25/2024] Open
Abstract
OBJECTIVE The purpose of this study is to investigate the role of high mobility group protein B1 (HMGB1) in placental development and fetal growth. METHODS We employed the Cre-loxP recombination system to establish a placenta-specific HMGB1 knockout mouse model. Breeding HMGB1flox/flox mice with Elf5-Cre mice facilitated the knockout, leveraging Elf5 expression in extra-embryonic ectoderm, ectoplacental cone, and trophoblast giant cells at 12.5 days of embryonic development. The primary goal of this model was to elucidate the molecular mechanism of HMGB1 in placental development, assessing parameters such as placental weight, fetal weight, and bone development. Additionally, we utilized lentiviral interference and overexpression of HMGB1 in human trophoblast cells to further investigate HMGB1's functional role. RESULTS Our findings indicate that the HMGB1flox/floxElf5cre/+ mouse displays fetal growth restriction, characterized by decreased placental and fetal weight and impaired bone development. The absence of HMGB1 inhibits autophagosome formation, impairs lysosomal degradation, and disrupts autophagic flux. Depletion of HMGB1 in human trophoblast cells also suppresses cell viability, proliferation, migration, and invasion by inhibiting the ERK signaling pathway. Overexpression of HMGB1 observed the opposite phenotypes. CONCLUSIONS HMGB1 participates in the regulation of autophagy through the ERK signaling pathway and affects placental development.
Collapse
Affiliation(s)
- Ming-Rui Li
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Toxicology, Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, China
| | - En-Xiang Chen
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Zhuo-Hang Li
- Department of Toxicology, Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Hong-Lan Song
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Zhang
- Department of Toxicology, Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Fang-Fang Li
- Department of Toxicology, Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, China
| | - You-Long Xie
- Department of Toxicology, Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Jing Tang
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Yu-Bin Ding
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Toxicology, Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Li-Juan Fu
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pharmacology, Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, School of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
- Department of Basic Medical Sciences, Changsha Medical University, Changsha, Hunan, China
| |
Collapse
|
3
|
Azmi MB, Nasir MF, Asif U, Kazi M, Uddin MN, Qureshi SA. Analyzing molecular signatures in preeclampsia and fetal growth restriction: Identifying key genes, pathways, and therapeutic targets for preterm birth. Front Mol Biosci 2024; 11:1384214. [PMID: 38712342 PMCID: PMC11070483 DOI: 10.3389/fmolb.2024.1384214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 03/22/2024] [Indexed: 05/08/2024] Open
Abstract
Background Intrauterine growth restriction (IUGR) and preeclampsia (PE) are intricately linked with specific maternal health conditions, exhibit shared placental abnormalities, and play pivotal roles in precipitating preterm birth (PTB) incidences. However, the molecular mechanism underlying the association between PE and IUGR has not been determined. Therefore, we aimed to analyze the data of females with PE and those with PE + IUGR to identify the key gene(s), their molecular pathways, and potential therapeutic interactions. Methods In this study, a comprehensive relationship analysis of both PE and PE + IUGR was conducted using RNA sequence datasets. Using two datasets (GSE148241 and GSE114691), differential gene expression analysis via DESeq2 through R-programming was performed. Gene set enrichment analysis was performed using ClusterProfiler, protein‒protein interaction (PPI) networks were constructed, and cluster analyses were conducted using String and MCODE in Cytoscape. Functional enrichment analyses of the resulting subnetworks were performed using ClueGO software. The hub genes were identified under both conditions using the CytoHubba method. Finally, the most common hub protein was docked against a library of bioactive flavonoids and PTB drugs using the PyRx AutoDock tool, followed by molecular dynamic (MD) simulation analysis. Pharmacokinetic analysis was performed to determine the ADMET properties of the compounds using pkCSM. Results We identified eight hub genes highly expressed in the case of PE, namely, PTGS2, ENG, KIT, MME, CGA, GAPDH, GPX3, and P4HA1, and the network of the PE + IUGR gene set demonstrated that nine hub genes were overexpressed, namely, PTGS2, FGF7, FGF10, IL10, SPP1, MPO, THBS1, CYBB, and PF4. PTGS2 was the most common hub gene found under both conditions (PE and PEIUGR). Moreover, the greater (-9.1 kcal/mol) molecular binding of flavoxate to PTGS2 was found to have satisfactory pharmacokinetic properties compared with those of other compounds. The flavoxate-bound PTGS2 protein complex remained stable throughout the simulation; with a ligand fit to protein, i.e., a RMSD ranging from ∼2.0 to 4.0 Å and a RMSF ranging from ∼0.5 to 2.9 Å, was observed throughout the 100 ns analysis. Conclusion The findings of this study may be useful for treating PE and IUGR in the management of PTB.
Collapse
Affiliation(s)
- Muhammad Bilal Azmi
- Computational Biochemistry Research Laboratory, Department of Biochemistry, Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Mushyeda Fatima Nasir
- Department of Biosciences, Faculty of Life Sciences, Mohammad Ali Jinnah University, Karachi, Pakistan
| | - Uzma Asif
- Department of Biochemistry, Medicine Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | | | | |
Collapse
|
4
|
Budal EB, Kessler J, Eide GE, Ebbing C, Collett K. Placental pathology and neonatal morbidity: exploring the impact of gestational age at birth. BMC Pregnancy Childbirth 2024; 24:201. [PMID: 38486145 PMCID: PMC10938777 DOI: 10.1186/s12884-024-06392-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 03/04/2024] [Indexed: 03/18/2024] Open
Abstract
AIM To evaluate placental pathology in term and post-term births, investigate differences in clinical characteristics, and assess the risk of adverse neonatal outcome. METHODS This prospective observational study included 315 singleton births with gestational age (GA) > 36 weeks + 6 days meeting the local criteria for referral to placental histopathologic examination. We applied the Amsterdam criteria to classify the placentas. Births were categorized according to GA; early-term (37 weeks + 0 days to 38 weeks + 6 days), term (39 weeks + 0 days to 40 weeks + 6 days), late-term (41 weeks + 0 days to 41 weeks + 6 days), and post-term births (≥ 42 weeks + 0 days). The groups were compared regarding placental pathology findings and clinical characteristics. Adverse neonatal outcomes were defined as 5-minute Apgar score < 7, umbilical cord artery pH < 7.0, admission to the neonatal intensive care unit or intrauterine death. A composite adverse outcome included one or more adverse outcomes. The associations between placental pathology, adverse neonatal outcomes, maternal and pregnancy characteristics were evaluated by logistic regression analysis. RESULTS Late-term and post-term births exhibited significantly higher rates of histologic chorioamnionitis (HCA), fetal inflammatory response, clinical chorioamnionitis (CCA) and transfer to neonatal intensive care unit (NICU) compared to early-term and term births. HCA and maternal smoking in pregnancy were associated with adverse outcomes in an adjusted analysis. Nulliparity, CCA, emergency section and increasing GA were all significantly associated with HCA. CONCLUSIONS HCA was more prevalent in late and post-term births and was the only factor, along with maternal smoking, that was associated with adverse neonatal outcomes. Since nulliparity, CCA and GA beyond term are associated with HCA, this should alert the clinician and elicit continuous intrapartum monitoring for timely intervention.
Collapse
Affiliation(s)
- Elisabeth B Budal
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Jørg Kessler
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Geir Egil Eide
- Centre for Clinical Research, Haukeland University Hospital, Bergen, Norway
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
- Western Norway University of Applied Sciences, Bergen, Norway
| | - Cathrine Ebbing
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Karin Collett
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.
- Department of Pathology, Haukeland University Hospital, Bergen, Norway.
- Department of Pathology, Helse Bergen HF, Haukeland University Hospital, Post box 1400, Bergen, N-5021, Norway.
| |
Collapse
|
5
|
Rahnavard A, Chatterjee R, Wen H, Gaylord C, Mugusi S, Klatt KC, Smith ER. Molecular epidemiology of pregnancy using omics data: advances, success stories, and challenges. J Transl Med 2024; 22:106. [PMID: 38279125 PMCID: PMC10821542 DOI: 10.1186/s12967-024-04876-7] [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: 02/09/2023] [Accepted: 12/26/2023] [Indexed: 01/28/2024] Open
Abstract
Multi-omics approaches have been successfully applied to investigate pregnancy and health outcomes at a molecular and genetic level in several studies. As omics technologies advance, research areas are open to study further. Here we discuss overall trends and examples of successfully using omics technologies and techniques (e.g., genomics, proteomics, metabolomics, and metagenomics) to investigate the molecular epidemiology of pregnancy. In addition, we outline omics applications and study characteristics of pregnancy for understanding fundamental biology, causal health, and physiological relationships, risk and prediction modeling, diagnostics, and correlations.
Collapse
Affiliation(s)
- Ali Rahnavard
- Computational Biology Institute, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA.
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA.
| | - Ranojoy Chatterjee
- Computational Biology Institute, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA
| | - Hui Wen
- Computational Biology Institute, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA
| | - Clark Gaylord
- Computational Biology Institute, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA
| | - Sabina Mugusi
- Department of Clinical Pharmacology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Kevin C Klatt
- Nutritional Sciences & Toxicology, University of California, Berkeley, CA, 94720, USA
| | - Emily R Smith
- Department of Global Health, The Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA.
- Department of Exercise and Nutrition Sciences, The Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA.
| |
Collapse
|
6
|
Mircheva A, Vangrieken P, Al-Nasiry S, van Schooten FJ, Godschalk RWL, Langie SAS. Optimizing the Comet Assay-Based In Vitro DNA Repair Assay for Placental Tissue: A Pilot Study with Pre-Eclamptic Patients. Int J Mol Sci 2023; 25:187. [PMID: 38203356 PMCID: PMC10779140 DOI: 10.3390/ijms25010187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
The comet assay-based in vitro DNA repair assay has become a common tool for quantifying base excision repair (BER) activity in human lymphocytes or cultured cells. Here, we optimized the protocol for studying BER in human placental tissue because the placenta is a non-invasive tissue for biomonitoring of early-life exposures, and it can be used to investigate molecular mechanisms associated with prenatal disorders. The optimal protein concentration of placental protein extracts for optimal damage recognition and incision was 2 mg protein/mL. The addition of aphidicolin did not lead to reduced non-specific incisions and was, therefore, not included in the optimized protocol. The interval between sample collection and analysis did not affect BER activity up to 70 min. Finally, this optimized protocol was tested on pre-eclamptic (PE) placental tissues (n = 11) and significantly lower BER activity in PE placentas compared to controls (n = 9) was observed. This was paralleled by a significant reduction in the expression of BER-related genes and increased DNA oxidation in PE placentas. Our study indicates that BER activity can be determined in placentas, and lower activity is present in PE compared with healthy. These findings should be followed up in prospective clinical investigations to examine BER's role in the advancement of PE.
Collapse
Affiliation(s)
- Anastasiya Mircheva
- Department of Pharmacology and Toxicology, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands; (A.M.); (R.W.L.G.)
| | - Philippe Vangrieken
- Department of Internal Medicine, School of Cardiovascular diseases (CARIM), Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Salwan Al-Nasiry
- Department of Obstetrics and Gynecology, Maastricht University Medical Center+, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| | - Frederik-Jan van Schooten
- Department of Pharmacology and Toxicology, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands; (A.M.); (R.W.L.G.)
| | - Roger W. L. Godschalk
- Department of Pharmacology and Toxicology, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands; (A.M.); (R.W.L.G.)
| | - Sabine A. S. Langie
- Department of Pharmacology and Toxicology, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands; (A.M.); (R.W.L.G.)
| |
Collapse
|
7
|
Spinillo A, Dominoni M, Mas FD, Cesari S, Fiandrino G, Gardella B. Placental fetal vascular malperfusion, neonatal neurologic morbidity, and infant neurodevelopmental outcomes: a systematic review and meta-analysis. Am J Obstet Gynecol 2023; 229:632-640.e2. [PMID: 37315755 DOI: 10.1016/j.ajog.2023.06.014] [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: 12/20/2022] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023]
Abstract
OBJECTIVE This study aimed to evaluate the association of placental fetal vascular malperfusion lesions with neonatal brain injury and adverse infant neurodevelopmental outcomes. DATA SOURCES PubMed and Medline, Scopus, and Cochrane databases were searched from inception to July 2022. STUDY ELIGIBILITY CRITERIA We included cohort and case-control studies reporting the associations of fetal vascular malperfusion lesions with neonatal encephalopathy, perinatal stroke, intracranial hemorrhage, periventricular leukomalacia, and infant neurodevelopmental and cognitive outcomes. METHODS Data were analyzed by including fetal vascular malperfusion lesions as an exposure variable and brain injuries or neurodevelopmental impairment as outcomes using random-effects models. The effect of moderators, such as gestational age or study type, was assessed by subgroup analysis. Study quality and risk of bias were assessed by applying the Observational Study Quality Evaluation method. RESULTS Out of the 1115 identified articles, 26 were selected for quantitative analysis. The rates of neonatal central nervous system injury (neonatal encephalopathy or perinatal stroke) in term or near-term infants were more common among fetal vascular malperfusion cases (n=145) than among controls (n=1623) (odds ratio, 4.00; 95% confidence interval, 2.72-5.90). In premature deliveries, fetal vascular malperfusion lesions did not influence the risk of intracranial hemorrhage or periventricular leukomalacia (odds ratio, 1.40; 95% confidence interval, 0.90-2.18). Fetal vascular malperfusion-associated risk of abnormal infant neurodevelopmental outcome (314 fetal vascular malperfusion cases and 1329 controls) was modulated by gestational age being higher in term infants (odds ratio, 5.02; 95% confidence interval, 1.59-15.91) than in preterm infants (odds ratio, 1.70; 95% confidence interval, 1.13-2.56). Abnormal infant cognitive development and mental development were more common among fetal vascular malperfusion cases (n=241) than among controls (n=2477) (odds ratio, 2.14; 95% confidence interval, 1.40-3.27). The type of study (cohort vs case-control) did not influence the association between fetal vascular malperfusion and subsequent infant brain injury or abnormal neurodevelopmental outcome. CONCLUSION The findings of cohort and case-control studies indicate a considerable association between fetal vascular malperfusion placental lesions and increased risk of brain injury in term neonates, and neurodevelopmental impairment in both term and preterm infants. A diagnosis of placental fetal vascular malperfusion should be taken into consideration by both pediatricians and neurologists during the follow-up of infants at risk of adverse neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Arsenio Spinillo
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Departments of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Mattia Dominoni
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Departments of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Francesca Dal Mas
- Department of Management, Ca' Foscari University of Venice, Venice, Italy
| | - Stefania Cesari
- Department of Pathology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giacomo Fiandrino
- Department of Pathology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Barbara Gardella
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Departments of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| |
Collapse
|
8
|
Kwok J, Speyer LG, Soursou G, Murray AL, Fanti KA, Auyeung B. Maternal metabolic syndrome in pregnancy and child development at age 5: exploring mediating mechanisms using cord blood markers. BMC Med 2023; 21:124. [PMID: 37013575 PMCID: PMC10071709 DOI: 10.1186/s12916-023-02835-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 03/15/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND There is limited evidence on how the classification of maternal metabolic syndrome during pregnancy affects children's developmental outcomes and the possible mediators of this association. This study uses a cohort sample of 12,644 to 13,832 mother-child pairs from the UK Born in Bradford Study to examine the associations between maternal metabolic syndrome classification (MetS) and child development outcomes at age 5, using cord blood markers as candidate mediators. METHODS Maternal cardiometabolic markers included diabetes, obesity, triglycerides, high-density lipoprotein cholesterol, blood pressure, hypertension, and fasting glucose during pregnancy. Cord blood markers of high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, triglycerides, leptin, and adiponectin were used as child mediators. Child outcomes included two starting school variables: British Picture Vocabulary Scale (BPVS) and the Letter Identification Assessment (LID), and five developmental milestone domains from a national UK framework: (1) communication and language (COM); (2) personal, social, and emotional (PSE); (3) physical development (PHY); (4) literacy (LIT); and (5) mathematics (MAT). Mediation models were used to examine the associations between the classification of maternal metabolic syndrome and child developmental milestones. Models were adjusted for potential maternal, socioeconomic, and child confounders such as maternal education, deprivation, and gestational age. RESULTS In mediation models, significant total effects were found for MetS associations with children's development in the LIT domain at age 5. MetS predicted individual cord blood mediators of lower HDL and increased leptin levels in both adjusted and unadjusted models. Total indirect effects (effects of all mediators combined) for MetS on a child's COM and PSE domain were significant, through all child cord blood mediators of LDL, HDL, triglycerides, adiponectin, and leptin for adjusted models. CONCLUSIONS The results support the hypothesis that maternal metabolic syndrome classification during pregnancy is associated with some child developmental outcomes at age 5. After adjusting for maternal, child, and environmental covariates, maternal metabolic syndrome classification during pregnancy was associated with children's LIT domain through direct effects of maternal metabolic health and indirect effects of cord blood markers (total effects), and COM and PSE domains via changes only in a child's cord blood markers (total indirect effects).
Collapse
Affiliation(s)
- Janell Kwok
- School of Philosophy, Psychology and Language Sciences, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK.
| | - Lydia Gabriela Speyer
- School of Philosophy, Psychology and Language Sciences, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Georgia Soursou
- Center for Applied Neuroscience, University of Cyprus, Nicosia, Cyprus
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
| | - Aja Louise Murray
- School of Philosophy, Psychology and Language Sciences, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
| | - Kostas A Fanti
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
| | - Bonnie Auyeung
- School of Philosophy, Psychology and Language Sciences, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
9
|
David AL, Spencer RN. Clinical Assessment of Fetal Well-Being and Fetal Safety Indicators. J Clin Pharmacol 2022; 62 Suppl 1:S67-S78. [PMID: 36106777 PMCID: PMC9544851 DOI: 10.1002/jcph.2126] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/24/2022] [Indexed: 12/03/2022]
Abstract
Delivering safe clinical trials of novel therapeutics is central to enable pregnant women and their babies to access medicines for better outcomes. This review describes clinical monitoring of fetal well-being and safety. Current pregnancy surveillance includes regular antenatal checks of blood pressure and urine for signs of gestational hypertension. Fetal and placental development is assessed routinely using the first-trimester "dating" and mid-trimester "anomaly" ultrasound scans, but the detection of fetal anomalies can continue throughout pregnancy using targeted sonography or magnetic resonance imaging (MRI). Serial sonography can be used to assess fetal size, well-being, and placental function. Carefully defined reproducible imaging parameters, such as the head circumference (HC), abdominal circumference (AC), and femur length (FL), are combined to calculate an estimate of the fetal weight. Doppler analysis of maternal uterine blood flow predicts placental insufficiency, which is associated with poor fetal growth. Fetal doppler analysis can indicate circulatory decompensation and fetal hypoxia, requiring delivery to be expedited. Novel ways to assess fetal well-being and placental function using MRI, computerized cardiotocography (CTG), serum circulating fetoplacental proteins, and mRNA may improve the assessment of the safety and efficacy of maternal and fetal interventions. Progress has been made in how to define and grade clinical trial safety in pregnant women, the fetus, and neonate. A new system for improved safety monitoring for clinical trials in pregnancy, Maternal and Fetal Adverse Event Terminology (MFAET), describes 12 maternal and 18 fetal adverse event (AE) definitions and severity grading criteria developed through an international modified Delphi consensus process. This fills a vital gap in maternal and fetal translational medicine research.
Collapse
Affiliation(s)
- Anna L. David
- Elizabeth Garrett Anderson Institute for Women's HealthUniversity College LondonLondonUK
- National Institute for Health and Care Research (NIHR) University College London Hospitals NHS Foundation Trust (UCLH)Biomedical Research CentreLondonUK
| | - Rebecca N. Spencer
- Elizabeth Garrett Anderson Institute for Women's HealthUniversity College LondonLondonUK
- School of MedicineUniversity of LeedsLeedsUK
| |
Collapse
|
10
|
Improving Development of Drug Treatments for Pregnant Women and the Fetus. Ther Innov Regul Sci 2022; 56:976-990. [PMID: 35881237 PMCID: PMC9315086 DOI: 10.1007/s43441-022-00433-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/30/2022] [Indexed: 12/12/2022]
Abstract
The exclusion of pregnant populations, women of reproductive age, and the fetus from clinical trials of therapeutics is a major global public health issue. It is also a problem of inequity in medicines development, as pregnancy is a protected characteristic. The current regulatory requirements for drugs in pregnancy are being analyzed by a number of agencies worldwide. There has been considerable investment in developing expertise in pregnancy clinical trials (for the pregnant person and the fetus) such as the Obstetric-Fetal Pharmacology Research Centers funded by the National Institute of Child Health and Human Development. Progress has also been made in how to define and grade clinical trial safety in pregnant women, the fetus, and neonate. Innovative methods to model human pregnancy physiology and pharmacology using computer simulations are also gaining interest. Novel ways to assess fetal well-being and placental function using magnetic resonance imaging, computerized cardiotocography, serum circulating fetoplacental proteins, and mRNA may permit better assessment of the safety and efficacy of interventions in the mother and fetus. The core outcomes in women’s and newborn health initiative is facilitating the consistent reporting of data from pregnancy trials. Electronic medical records integrated with pharmacy services should improve the strength of pharmacoepidemiologic and pharmacovigilance studies. Incentives such as investigational plans and orphan disease designation have been taken up for obstetric, fetal, and neonatal diseases. This review describes the progress that is being made to better understand the extent of the problem and to develop applicable solutions.
Collapse
|
11
|
The Update of Fetal Growth Restriction Associated with Biomarkers. MATERNAL-FETAL MEDICINE 2022. [DOI: 10.1097/fm9.0000000000000156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
12
|
Jackson WM, Santos HP, Hartwell HJ, Gower WA, Chhabra D, Hagood JS, Laughon MM, Payton A, Smeester L, Roell K, O’Shea TM, Fry RC. Differential placental CpG methylation is associated with chronic lung disease of prematurity. Pediatr Res 2022; 91:1428-1435. [PMID: 34857876 PMCID: PMC9160210 DOI: 10.1038/s41390-021-01868-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 10/26/2021] [Accepted: 11/13/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Chronic lung disease (CLD) is the most common pulmonary morbidity in extremely preterm infants. It is unclear to what extent prenatal exposures influence the risk of CLD. Epigenetic variation in placenta DNA methylation may be associated with differential risk of CLD, and these associations may be dependent upon sex. METHODS Data were obtained from a multi-center cohort of infants born extremely preterm (<28 weeks' gestation) and an epigenome-wide approach was used to identify associations between placental DNA methylation and CLD (n = 423). Associations were evaluated using robust linear regression adjusting for covariates, with a false discovery rate of 0.05. Analyses stratified by sex were used to assess differences in methylation-CLD associations. RESULTS CLD was associated with differential methylation at 49 CpG sites representing 46 genes in the placenta. CLD was associated with differential methylation of probes within genes related to pathways involved in fetal lung development, such as p53 signaling and myo-inositol biosynthesis. Associations between CpG methylation and CLD differed by sex. CONCLUSIONS Differential placental methylation within genes with key roles in fetal lung development may reflect complex cell signaling between the placenta and fetus which mediate CLD risk. These pathways appear to be distinct based on fetal sex. IMPACT In extremely preterm infants, differential methylation of CpG sites within placental genes involved in pathways related to cell signaling, oxidative stress, and trophoblast invasion is associated with chronic lung disease of prematurity. DNA methylation patterns associated with chronic lung disease were distinctly based on fetal sex, suggesting a potential mechanism underlying dimorphic phenotypes. Mechanisms related to fetal hypoxia and placental myo-inositol signaling may play a role in fetal lung programming and the developmental origins of chronic lung disease. Continued research of the relationship between the placental epigenome and chronic lung disease could inform efforts to ameliorate or prevent this condition.
Collapse
Affiliation(s)
- Wesley M. Jackson
- Department of Pediatrics, School of Medicine, The University of North Carolina, Chapel Hill, NC
| | - Hudson P. Santos
- Department of Environmental Sciences and Engineering, Gilling School of Global Public Health, The University of North Carolina, Chapel Hill, NC.,Biobehavioral Laboratory, School of Nursing, The University of North Carolina, Chapel Hill, NC
| | - Hadley J. Hartwell
- Department of Environmental Sciences and Engineering, Gilling School of Global Public Health, The University of North Carolina, Chapel Hill, NC
| | - William Adam Gower
- Department of Pediatrics, School of Medicine, The University of North Carolina, Chapel Hill, NC
| | - Divya Chhabra
- Department of Pediatrics, University of California, San Diego, CA
| | - James S. Hagood
- Department of Pediatrics, School of Medicine, The University of North Carolina, Chapel Hill, NC
| | - Matthew M. Laughon
- Department of Pediatrics, School of Medicine, The University of North Carolina, Chapel Hill, NC
| | - Alexis Payton
- Department of Environmental Sciences and Engineering, Gilling School of Global Public Health, The University of North Carolina, Chapel Hill, NC.,Institute for Environmental Health Solutions, Gilling School of Global Public Health, The University of North Carolina, Chapel Hill, NC
| | - Lisa Smeester
- Department of Environmental Sciences and Engineering, Gilling School of Global Public Health, The University of North Carolina, Chapel Hill, NC.,Institute for Environmental Health Solutions, Gilling School of Global Public Health, The University of North Carolina, Chapel Hill, NC
| | - Kyle Roell
- Department of Environmental Sciences and Engineering, Gilling School of Global Public Health, The University of North Carolina, Chapel Hill, NC.,Institute for Environmental Health Solutions, Gilling School of Global Public Health, The University of North Carolina, Chapel Hill, NC
| | - T. Michael O’Shea
- Department of Pediatrics, School of Medicine, The University of North Carolina, Chapel Hill, NC
| | - Rebecca C. Fry
- Department of Environmental Sciences and Engineering, Gilling School of Global Public Health, The University of North Carolina, Chapel Hill, NC.,Institute for Environmental Health Solutions, Gilling School of Global Public Health, The University of North Carolina, Chapel Hill, NC
| |
Collapse
|
13
|
Benagiano M, Mancuso S, Brosens JJ, Benagiano G. Long-Term Consequences of Placental Vascular Pathology on the Maternal and Offspring Cardiovascular Systems. Biomolecules 2021; 11:1625. [PMID: 34827623 PMCID: PMC8615676 DOI: 10.3390/biom11111625] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/10/2021] [Accepted: 10/13/2021] [Indexed: 12/14/2022] Open
Abstract
Over the last thirty years, evidence has been accumulating that Hypertensive Disorders of Pregnancy (HDP) and, specifically, Preeclampsia (PE) produce not only long-term effects on the pregnant woman, but have also lasting consequences for the fetus. At the core of these consequences is the phenomenon known as defective deep placentation, being present in virtually every major obstetrical syndrome. The profound placental vascular lesions characteristic of this pathology can induce long-term adverse consequences for the pregnant woman's entire arterial system. In addition, placental growth restriction and function can, in turn, cause a decreased blood supply to the fetus, with long-lasting effects. Women with a history of HDP have an increased risk of Cardiovascular Diseases (CVD) compared with women with normal pregnancies. Specifically, these subjects are at a future higher risk of: Hypertension; Coronary artery disease; Heart failure; Peripheral vascular disease; Cerebrovascular accidents (Stroke); CVD-related mortality. Vascular pathology in pregnancy and CVD may share a common etiology and may have common risk factors, which are unmasked by the "stress" of pregnancy. It is also possible that the future occurrence of a CVD may be the consequence of endothelial dysfunction generated by pregnancy-induced hypertension that persists after delivery. Although biochemical and biophysical markers of PE abound, information on markers for a comparative evaluation in the various groups is still lacking. Long-term consequences for the fetus are an integral part of the theory of a fetal origin of a number of adult diseases, known as the Barker hypothesis. Indeed, intrauterine malnutrition and fetal growth restriction represent significant risk factors for the development of chronic hypertension, diabetes, stroke and death from coronary artery disease in adults. Other factors will also influence the development later in life of hypertension, coronary and myocardial disease; they include parental genetic disposition, epigenetic modifications, endothelial dysfunction, concurrent intrauterine exposures, and the lifestyle of the affected individual.
Collapse
Affiliation(s)
- Marisa Benagiano
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Salvatore Mancuso
- Department of Life Sciences, Catholic University of Rome, 00168 Rome, Italy;
| | - Jan J. Brosens
- Division of Biomedical Sciences, Warwick Medical School, Coventry CV4 7HL, UK;
| | - Giuseppe Benagiano
- Department of Maternal and Child Health, Gynecology and Urology, Sapienza University of Rome, 00185 Rome, Italy;
| |
Collapse
|
14
|
Andescavage N, Kapse K, Lu YC, Barnett SD, Jacobs M, Gimovsky AC, Ahmadzia H, Quistorff J, Lopez C, Andersen NR, Bulas D, Limperopoulos C. Normative placental structure in pregnancy using quantitative Magnetic Resonance Imaging. Placenta 2021; 112:172-179. [PMID: 34365206 DOI: 10.1016/j.placenta.2021.07.296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/08/2021] [Accepted: 07/27/2021] [Indexed: 11/29/2022]
Abstract
INTRODUCTION To characterize normative morphometric, textural and microstructural placental development by applying advanced and quantitative magnetic resonance imaging (qMRI) techniques to the in-vivo placenta. METHODS We enrolled 195 women with uncomplicated, healthy singleton pregnancies in a prospective observational study. Women underwent MRI between 16- and 40-weeks' gestation. Morphometric and textural metrics of placental growth were calculated from T2-weighted (T2W) images, while measures of microstructural development were calculated from diffusion-weighted images (DWI). Normative tables and reference curves were constructed for each measured index across gestation and according to fetal sex. RESULTS Data from 269 MRI studies from 169 pregnant women were included in the analyses. During the study period, placentas undergo significant increases in morphometric measures of volume, thickness, and elongation. Placental texture reveals increasing variability with advancing gestation as measured by grey level non uniformity, run length non uniformity and long run high grey level emphasis. Placental microstructure did not vary with gestational age. Placental elongation was the only metric that differed significantly between male and female fetuses. DISCUSSION We report quantitative metrics of placental morphometry, texture and microstructure in a large cohort of healthy controls during the second and third trimesters of pregnancy. These measures can serve as normative references of in-vivo placental development to better understand placental function in high-risk conditions and allow for the early detection of placental mal-development.
Collapse
Affiliation(s)
- Nickie Andescavage
- Division of Neonatology, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA; Department of Pediatrics, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Kushal Kapse
- Division of Diagnostic Imaging & Radiology, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Yuan-Chiao Lu
- Division of Diagnostic Imaging & Radiology, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Scott D Barnett
- Division of Diagnostic Imaging & Radiology, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Marni Jacobs
- Division of Biostatistics & Study Methodology, George Washington University School of Medicine, 2300 Eye St. NW, Washington, DC, 20037, USA
| | - Alexis C Gimovsky
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynecology, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Homa Ahmadzia
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynecology, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Jessica Quistorff
- Division of Diagnostic Imaging & Radiology, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Catherine Lopez
- Division of Diagnostic Imaging & Radiology, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Nicole Reinholdt Andersen
- Division of Diagnostic Imaging & Radiology, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Dorothy Bulas
- Division of Diagnostic Imaging & Radiology, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA; Department of Radiology, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA
| | - Catherine Limperopoulos
- Division of Diagnostic Imaging & Radiology, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA; Department of Pediatrics, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA; Department of Radiology, Children's National Hospital, 111 Michigan Ave. NW, Washington, DC, 20010, USA.
| |
Collapse
|
15
|
Marinello WP, Patisaul HB. Endocrine disrupting chemicals (EDCs) and placental function: Impact on fetal brain development. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 92:347-400. [PMID: 34452690 DOI: 10.1016/bs.apha.2021.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Pregnancy is a critical time of vulnerability for the development of the fetal brain. Exposure to environmental pollutants at any point in pregnancy can negatively impact many aspects of fetal development, especially the organization and differentiation of the brain. The placenta performs a variety of functions that can help protect the fetus and sustain brain development. However, disruption of any of these functions can have negative impacts on both the pregnancy outcome and fetal neurodevelopment. This review presents current understanding of how environmental exposures, specifically to endocrine disrupting chemicals (EDCs), interfere with placental function and, in turn, neurodevelopment. Some of the key differences in placental development between animal models are presented, as well as how placental functions such as serving as a xenobiotic barrier and exchange organ, immune interface, regulator of growth and fetal oxygenation, and a neuroendocrine organ, could be vulnerable to environmental exposure. This review illustrates the importance of the placenta as a modulator of fetal brain development and suggests critical unexplored areas and possible vulnerabilities to environmental exposure.
Collapse
Affiliation(s)
- William P Marinello
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States
| | - Heather B Patisaul
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States.
| |
Collapse
|
16
|
Trongpisutsak A, Phupong V. Prediction of preeclampsia using a combination of serum micro RNA-210 and uterine artery Doppler ultrasound. Sci Prog 2021; 104:368504211036856. [PMID: 34432986 PMCID: PMC10450716 DOI: 10.1177/00368504211036856] [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] [Indexed: 11/16/2022]
Abstract
The objective was to determine whether a combination of serum micro RNA-210 level and uterine artery Doppler can predict preeclampsia in pregnant women at 16-24 weeks gestation. A prospective observational study conducted in singleton pregnant women at 16-24 weeks of gestation who had prenatal care at the King Chulalongkorn Memorial Hospital, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand between 2017 and 2018. Uterine artery Doppler ultrasound and blood testing for serum micro RNA-210 were performed. Pregnancy outcomes were recorded. Optimal cut-off for uterine artery pulsatility index (PI) and serum micro RNA-210 were obtained to calculate the predictive values for preeclampsia. Data from 443 participants were analyzed. Twenty-two cases developed preeclampsia (5.0%) and seven of these preeclamptic cases had early-onset preeclampsia (1.6%). Pregnant women with preeclampsia had higher mean PI of the uterine artery (1.34 ± 0.52 vs 0.98 ± 0.28, p = 0.004), higher detection rates of diastolic notching (45.5% vs 11.2%, p < 0.001), and lower median serum micro RNA-210 level (22.86 vs 795.78, p < 0.001) than pregnant women without preeclampsia. Using abnormal serum micro RNA-210 level, abnormal mean PI or uterine artery diastolic notches to predict for preeclampsia, the sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) were 95.5%, 54.9%, 10.0%, and 99.6%, respectively. For early-onset preeclampsia prediction, the sensitivity, specificity, PPV, and NPV were 100.0%, 53.2%, 3.3%, and 100.0%, respectively. This study demonstrated that a combination of serum micro RNA-210 and uterine artery Doppler is effective in predicting preeclampsia in the second trimester.
Collapse
Affiliation(s)
- Ananya Trongpisutsak
- Placental Related Diseases Research Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Vorapong Phupong
- Placental Related Diseases Research Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
17
|
Groleau M, White F, Cardenas A, Perron P, Hivert MF, Bouchard L, Jacques PÉ. Comparative epigenome-wide analysis highlights placenta-specific differentially methylated regions. Epigenomics 2021; 13:357-368. [PMID: 33661023 DOI: 10.2217/epi-2020-0271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aim: The placenta undergoes DNA methylation (DNAm) programming that is unique compared with all other fetal tissues. We aim to decipher some of the physiologic roles of the placenta by comparing its DNAm profile with that of another fetal tissue. Materials & methods: We performed a comparative analysis of genome-wide DNAm of 444 placentas paired with cord blood samples collected at birth. Gene ontology term analyses were conducted on the resulting differentially methylated regions. Results: Genomic regions upstream of transcription start sites showing lower DNAm in the placenta were enriched with terms related to miRNA functions and genes encoding G-protein-coupled receptors. Conclusion: These results highlight genomic regions that are differentially methylated in the placenta in contrast to fetal blood.
Collapse
Affiliation(s)
- Marika Groleau
- Département de Biologie, Université de Sherbrooke, Sherbrooke, Québec, J1K 2R1, Canada
| | - Frédérique White
- Département de Biologie, Université de Sherbrooke, Sherbrooke, Québec, J1K 2R1, Canada
| | - Andres Cardenas
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, 94720-7360, USA
| | - Patrice Perron
- Département de Médecine, Université de Sherbrooke, Sherbrooke, Québec, J1K 2R1, Canada.,Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada
| | - Marie-France Hivert
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada.,Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA, 02115, USA.,Diabetes Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Luigi Bouchard
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada.,Department of Biochemistry & Functional Genomics, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada.,Department of Medical Biology, CIUSSS Saguenay-Lac-Saint-Jean, Hôpital de Chicoutimi, Saguenay, Québec, G7H 7K9, Canada
| | - Pierre-Étienne Jacques
- Département de Biologie, Université de Sherbrooke, Sherbrooke, Québec, J1K 2R1, Canada.,Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada
| |
Collapse
|
18
|
NIH Workshop 2018: Towards Minimally Invasive or Noninvasive Approaches to Assess Tissue Oxygenation Pre- and Post-transfusion. Transfus Med Rev 2020; 35:46-55. [PMID: 33353783 DOI: 10.1016/j.tmrv.2020.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/12/2022]
|
19
|
Vos S, Nawrot TS, Martens DS, Byun HM, Janssen BG. Mitochondrial DNA methylation in placental tissue: a proof of concept study by means of prenatal environmental stressors. Epigenetics 2020; 16:121-131. [PMID: 32657220 PMCID: PMC7889149 DOI: 10.1080/15592294.2020.1790923] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
While previous studies have demonstrated that prenatal exposure to environmental stressors is associated with mitochondrial DNA (mtDNA) methylation, more recent investigations are questioning the accuracy of the methylation assessment and its biological relevance. In this study, we investigated placental mtDNA methylation while accounting for methodological issues such as nuclear contamination, bisulphite conversion, and PCR bias. From the ENVIRONAGE birth cohort, we selected three groups of participants (n = 20/group). One group with mothers who smoked during pregnancy (average 13.2 cig/day), one group with high air pollutant exposure (PM2.5: 16.0 ± 1.4 µg/m3, black carbon: 1.8 ± 0.3 µg/m3) and one control group (non-smokers, PM2.5: 10.6 ± 1.7 µg/m3, black carbon: 0.9 ± 0.1 µg/m3) with low air pollutant exposure. DNA methylation levels were quantified in two regions of the displacement loop control region (D-loop and LDLR2) by bisulphite pyrosequencing. Additionally, we measured DNA methylation on nuclear genes involved in mitochondrial maintenance (PINK1, DNA2, and POLG1) and assessed mtDNA content using qPCR. Absolute D-loop methylation levels were higher for mothers that smoked extensively (+0.36%, 95% CI: 0.06% to 0.66%), and for mothers that were highly exposed to air pollutants (+0.47%, 95% CI: 0.20% to 0.73%). The relevance of our findings is further supported, as D-loop methylation levels were correlated with placental mtDNA content (r = −0.40, p = 0.002) and associated with birth weight (−106.98 g, 95% CI: −209.60 g to −4.36 g for an IQR increase in D-loop methylation). Most notably, our data demonstrates relevant levels of mtDNA methylation in placenta tissue, with significant associations between prenatal exposure to environmental stressors and D-loop methylation.
Collapse
Affiliation(s)
- Stijn Vos
- Centre for Environmental Sciences, Hasselt University , Hasselt, Belgium
| | - Tim S Nawrot
- Centre for Environmental Sciences, Hasselt University , Hasselt, Belgium.,Department of Public Health & Primary Care, Occupational & Environmental Medicine, Leuven University , Leuven, Belgium
| | - Dries S Martens
- Centre for Environmental Sciences, Hasselt University , Hasselt, Belgium
| | - Hyang-Min Byun
- Population Health Sciences Institute, Newcastle University , Newcastle upon Tyne, UK
| | - Bram G Janssen
- Centre for Environmental Sciences, Hasselt University , Hasselt, Belgium
| |
Collapse
|
20
|
Bonell A, Hirst J, Vicedo-Cabrera AM, Haines A, Prentice AM, Maxwell NS. A protocol for an observational cohort study of heat strain and its effect on fetal wellbeing in pregnant farmers in The Gambia. Wellcome Open Res 2020; 5:32. [PMID: 32292825 PMCID: PMC7141168 DOI: 10.12688/wellcomeopenres.15731.2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2020] [Indexed: 11/20/2022] Open
Abstract
Introduction: Climate change predictions indicate that global temperatures are likely to exceed those seen in the last 200,000 years, rising by around 4°C above pre-industrial levels by 2100 (without effective mitigation of current emission rates). In regions of the world set to experience extreme temperatures, women often work outside in agriculture even during pregnancy. The implications of heat strain in pregnancy on maternal health and pregnancy outcome are not well understood. This protocol describes a study to assess the physiological response of pregnant women to environmental heat stress and the immediate effect this has on fetal wellbeing. Methods and analysis: The study will be performed in West Kiang district, The Gambia; a semi-arid zone in West Africa with daily maximum temperatures ranging from approximately 32 to 40°C. We will recruit 125 pregnant women of all ages who perform agricultural work during their pregnancy. Participants will be followed every two months until delivery. At each study visit fetal growth will be measured by ultrasound scan. During the course of their working day we will take the following measurements: continuous maternal physiological measurements (heart rate, respiratory rate, chest skin temperature and tri-axis accelerometer data); intermittent maternal tympanic core temperature, four point skin temperature, blood pressure; intermittent fetal heart rate and, if eligible, umbilical artery doppler; intermittent environmental measurements of air temperature, humidity, solar radiation and wind speed. Venous blood and urine will be collected at beginning and end of day for biomarkers of heat strain or fetal distress and hydration status.
Collapse
Affiliation(s)
- Ana Bonell
- Medical Research Council Gambia @ London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Jane Hirst
- Nuffield Department of Women's and Reproductive Health and the George Institute for Global Health, University of Oxford, Oxford, UK
| | - Ana M Vicedo-Cabrera
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland.,Oeschger Center for Climate Change Research, University of Bern, Bern, Switzerland
| | - Andy Haines
- Department of Public Health, Environment and Society; Department of Population health, London School of Hygiene and Tropical Medicine, London, UK
| | - Andrew M Prentice
- Medical Research Council Gambia @ London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Neil S Maxwell
- Environmental Extremes Laboratory, University of Brighton, Brighton, UK
| |
Collapse
|
21
|
Bonell A, Hirst J, Vicedo-Cabrera AM, Haines A, Prentice AM, Maxwell NS. A protocol for an observational cohort study of heat strain and its effect on fetal wellbeing in pregnant farmers in The Gambia. Wellcome Open Res 2020; 5:32. [PMID: 32292825 PMCID: PMC7141168 DOI: 10.12688/wellcomeopenres.15731.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2020] [Indexed: 11/15/2023] Open
Abstract
Introduction: Climate change predictions indicate that global temperatures are likely to exceed those seen in the last 200,000 years, rising by around 4°C above pre-industrial levels by 2100 (without effective mitigation of current emission rates). In regions of the world set to experience extreme temperatures, women often work outside in agriculture even during pregnancy. The implications of heat strain in pregnancy on maternal health and pregnancy outcome are not well understood. This protocol describes a study to assess the physiological response of pregnant women to environmental heat stress and the immediate effect this has on fetal wellbeing. Methods and analysis: The study will be performed in West Kiang district, The Gambia; a semi-arid zone in West Africa with daily maximum temperatures ranging from approximately 32 to 40°C. We will recruit 125 pregnant women of all ages who perform agricultural work during their pregnancy. Participants will be followed every two months until delivery. At each study visit fetal growth will be measured by ultrasound scan. During the course of their working day we will take the following measurements: continuous maternal physiological measurements (heart rate, respiratory rate, chest skin temperature and tri-axis accelerometer data); intermittent maternal tympanic core temperature, four point skin temperature, blood pressure; intermittent fetal heart rate and, if eligible, umbilical artery doppler; intermittent environmental measurements of air temperature, humidity, solar radiation and wind speed. Venous blood and urine will be collected at beginning and end of day for biomarkers of heat strain or fetal distress and hydration status.
Collapse
Affiliation(s)
- Ana Bonell
- Medical Research Council Gambia @ London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Jane Hirst
- Nuffield Department of Women's and Reproductive Health and the George Institute for Global Health, University of Oxford, Oxford, UK
| | - Ana M. Vicedo-Cabrera
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Oeschger Center for Climate Change Research, University of Bern, Bern, Switzerland
| | - Andy Haines
- Department of Public Health, Environment and Society; Department of Population health, London School of Hygiene and Tropical Medicine, London, UK
| | - Andrew M. Prentice
- Medical Research Council Gambia @ London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Neil S. Maxwell
- Environmental Extremes Laboratory, University of Brighton, Brighton, UK
| |
Collapse
|
22
|
El-Samahy MA, Yao X, Zhang G, Zhang Y, Wang F. A proposed sample handling of ovine cotyledon for proteomic studies. Anal Biochem 2020; 593:113585. [PMID: 31954698 DOI: 10.1016/j.ab.2020.113585] [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: 09/16/2019] [Revised: 01/12/2020] [Accepted: 01/14/2020] [Indexed: 11/28/2022]
Abstract
Ovine trophoblast is a suitable material for placental studies. However, a universal protocol for handling ruminant cotyledon samples has still not been reported. Considering the villous structure of ovine cotyledon, we suggest procedures to prepare cotyledons with limited inherent contamination, using semi-dry conditions to avoid freezing damage and sample errors. The cytosolic water-soluble proteins were physically extracted from the frozen cotyledons. High homogeneity was demonstrated between the replicates of both tissue and extract samples. Importantly, the chemical lysis of placental crude extracts was necessary for protein separation and immunoreaction. The integrity of stored tissues was histologically validated using a formalin-fixed paraffin-embedded technique. Using label-free proteomics, we detected 388 Ovis protein-groups in at least two of three biological replicates of either the tissue or extract. Although the water-soluble proteins were dominated by hemoglobin subunits, ten proteins were identified exclusively in all extract replicates. The physical extraction selectively reduced the membrane, extracellular matrix, and cytoskeleton proteins. The hydrolase enzymes, in the extract, hindered the identification of some specific proteins, such as histone H2A. In summary, the proposed workflow may guide further proteomic investigations of ovine cotyledon biology. Furthermore, our proteomic data have inferred some potential mechanisms of ovine trophoblast at parturition.
Collapse
Affiliation(s)
- M A El-Samahy
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China
| | - Xiaolei Yao
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China
| | - Guomin Zhang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China; Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China
| | - Yanli Zhang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China
| | - Feng Wang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China.
| |
Collapse
|
23
|
Konwar C, Del Gobbo G, Yuan V, Robinson WP. Considerations when processing and interpreting genomics data of the placenta. Placenta 2019; 84:57-62. [PMID: 30642669 PMCID: PMC6612459 DOI: 10.1016/j.placenta.2019.01.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/28/2018] [Accepted: 01/04/2019] [Indexed: 12/27/2022]
Abstract
The application of genomic approaches to placental research has opened exciting new avenues to help us understand basic biological properties of the placenta, improve prenatal screening/diagnosis, and measure effects of in utero exposures on child health outcomes. In the last decade, such large-scale genomic data (including epigenomics and transcriptomics) have become more easily accessible to researchers from many disciplines due to the increasing ease of obtaining such data and the rapidly evolving computational tools available for analysis. While the potential of large-scale studies has been widely promoted, less attention has been given to some of the challenges associated with processing and interpreting such data. We hereby share some of our experiences in assessing data quality, reproducibility, and interpretation in the context of genome-wide studies of the placenta, with the aim to improve future studies. There is rarely a single "best" approach, as that can depend on the study question and sample cohort. However, being consistent, thoroughly assessing potential confounders in the data, and communicating key variables in the methods section of the manuscript are critically important to help researchers to collaborate and build on each other's work.
Collapse
Affiliation(s)
- Chaini Konwar
- BC Children's Hospital Research Institute, 950 West 28th Ave, Vancouver, BC V5Z 4H4, Canada; Department of Medical Genetics, University of British Columbia, 4500, Oak Street, Vancouver, BC V6H3N1, Canada.
| | - Giulia Del Gobbo
- BC Children's Hospital Research Institute, 950 West 28th Ave, Vancouver, BC V5Z 4H4, Canada; Department of Medical Genetics, University of British Columbia, 4500, Oak Street, Vancouver, BC V6H3N1, Canada.
| | - Victor Yuan
- BC Children's Hospital Research Institute, 950 West 28th Ave, Vancouver, BC V5Z 4H4, Canada; Department of Medical Genetics, University of British Columbia, 4500, Oak Street, Vancouver, BC V6H3N1, Canada.
| | - Wendy P Robinson
- BC Children's Hospital Research Institute, 950 West 28th Ave, Vancouver, BC V5Z 4H4, Canada; Department of Medical Genetics, University of British Columbia, 4500, Oak Street, Vancouver, BC V6H3N1, Canada.
| |
Collapse
|
24
|
Yamaguchi Y, Tayama C, Tomikawa J, Akaishi R, Kamura H, Matsuoka K, Wake N, Minakami H, Kato K, Yamada T, Nakabayashi K, Hata K. Placenta-specific epimutation at H19-DMR among common pregnancy complications: its frequency and effect on the expression patterns of H19 and IGF2. Clin Epigenetics 2019; 11:113. [PMID: 31370882 PMCID: PMC6676526 DOI: 10.1186/s13148-019-0712-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 07/22/2019] [Indexed: 12/17/2022] Open
Abstract
Background H19 and IGF2 genes are imprinted and involved in regulating fetal and placental growth. The H19 differentially methylated region (DMR) is paternally methylated and maternally unmethylated and regulates the imprinted expression of H19 and IGF2. Epimutation at the H19-DMR in humans results in congenital growth disorders, Beckwith-Wiedemann and Silver-Russell syndromes, when erroneously its maternal allele becomes methylated and its paternal allele becomes unmethylated, respectively. Although H19 and IGF2 have been assessed for their involvement in pregnancy complications including fetal growth restriction (FGR) and pregnancy-induced hypertension (PIH)/hypertensive disorder of pregnancy (HDP) intensively in the last decade, it is still not established whether epimutation at the H19-DMR in the placenta results in pathogenic conditions in pregnancy. We aimed to assess the frequency of H19-DMR epimutation and its effects on the allelic expression patterns of H19 and IGF2 genes among normal and abnormal pregnancy cases. Results We enrolled two independently collected sets of placenta samples from normal pregnancies as controls and common pregnancy complications, FGR and PIH (HDP). The first set consisted of 39 controls and 140 FGR and/or PIH cases, and the second set consisted of 29 controls and 62 cases. For these samples, we initially screened for DNA methylation changes at H19-DMR and IGF2-DMRs by combined bisulfite restriction analysis, and further analyzed cases with methylation changes for their allelic methylation and expression patterns. We identified one case each of FGR and PIH showing hypomethylation of H19-DMR and IGF2-DMRs only in the placenta, but not in cord blood, from the first case/control set. For the PIH case, we were able to determine the allelic expression pattern of H19 to be biallelically expressed and the H19/IGF2 expression ratio to be highly elevated compared to controls. We also identified a PIH case with hypomethylation at H19-DMR and IGF2-DMRs in the placenta from the second case/control set. Conclusions Placental epimutation at H19-DMR was observed among common pregnancy complication cases at the frequency of 1.5% (3 out of 202 cases examined), but not in 68 normal pregnancy cases examined. Alteration of H19/IGF2 expression patterns due to hypomethylation of H19-DMR may have been involved in the pathogenesis of pregnancy complications in these cases. Electronic supplementary material The online version of this article (10.1186/s13148-019-0712-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuko Yamaguchi
- Department of Maternal-Fetal Biology, Research Institute, National Center for Child Health and Development, Tokyo, 157-8535, Japan.,Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Chiharu Tayama
- Department of Maternal-Fetal Biology, Research Institute, National Center for Child Health and Development, Tokyo, 157-8535, Japan
| | - Junko Tomikawa
- Department of Maternal-Fetal Biology, Research Institute, National Center for Child Health and Development, Tokyo, 157-8535, Japan
| | - Rina Akaishi
- Center of Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo, 157-8535, Japan
| | - Hiromi Kamura
- Department of Maternal-Fetal Biology, Research Institute, National Center for Child Health and Development, Tokyo, 157-8535, Japan
| | - Kentaro Matsuoka
- Department of Pathology, National Center for Child Health and Development, Tokyo, 157-8535, Japan.,Present Address: Department of Pathology, Dokkyo Medical University, Saitama Medical Center, Koshigaya, Japan
| | - Norio Wake
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Hisanori Minakami
- Department of Obstetrics and Gynecology, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638, Japan
| | - Kiyoko Kato
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Takahiro Yamada
- Clinical Genetics Unit, Kyoto University Hospital, Kyoto, 606-8507, Japan
| | - Kazuhiko Nakabayashi
- Department of Maternal-Fetal Biology, Research Institute, National Center for Child Health and Development, Tokyo, 157-8535, Japan.
| | - Kenichiro Hata
- Department of Maternal-Fetal Biology, Research Institute, National Center for Child Health and Development, Tokyo, 157-8535, Japan.
| |
Collapse
|
25
|
Johnsen JM, Brown DL. The national blueprint for pregnancy/birth longitudinal cohorts to study factor VIII immunogenicity: NHLBI State of the Science (SOS) Workshop on factor VIII inhibitors. Haemophilia 2019; 25:603-609. [PMID: 31329365 DOI: 10.1111/hae.13739] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/03/2019] [Accepted: 02/21/2019] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Patients with haemophilia can develop inhibitors to exogenous coagulation factors. Some patients are tolerant to factor, while those who develop inhibitors do so early in life. Genetics and environmental factors are known to contribute to inhibitor risk. However, it is not yet possible to predict inhibitor formation or treatment responsiveness in individuals. We hypothesize that factors in the antenatal/neonatal period inform inhibitor risk development. AIM To consider the design of longitudinal studies beginning in the antenatal/neonatal period and the use of new technologies to better understand haemophilia inhibitors. METHODS A working group was formed for the NHLBI State of the Science Workshop: Factor VIII Inhibitors: Generating a National Blueprint for Future Research to solicit input from the US haemophilia community and international collaborators to consider design of pregnancy/birth longitudinal cohorts that leverage -omics, existing phenotypic data, and in silico modelling to study inhibitors. RESULTS An antenatal/neonatal longitudinal cohort should begin with enrolment of pregnant genetic carriers of haemophilia and span the at-risk period for inhibitor development in the child. Data and samples from the mother, placenta, neonate and young child can be obtained that are amenable to existing assays, genomics and other -omics studies. Data can inform in silico prediction and mathematical models. CONCLUSION A longitudinal study beginning before birth offers the unique opportunity to study factors that influence inhibitor development prior to exposure. Advances in -omics and computational biology can study complex phenotypes in this rare disease. This study could be accomplished through interdisciplinary efforts and patient community engagement.
Collapse
Affiliation(s)
- Jill M Johnsen
- Bloodworks Northwest Research Institute, Seattle, Washington.,Washington Center for Bleeding Disorders, Seattle, Washington.,Department of Medicine, University of Washington, Seattle, Washington
| | - Deborah L Brown
- University of Texas Health Science Center, Houston, Texas.,MD Anderson Cancer Center, Houston, Texas.,Gulf States Hemophilia and Thrombophilia Treatment Center, Houston, Texas
| | | |
Collapse
|
26
|
Maternal physical activity-induced adaptive transcriptional response in brain and placenta of mothers and rat offspring. J Dev Orig Health Dis 2019; 11:108-117. [PMID: 31203831 DOI: 10.1017/s2040174419000333] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Maternal physical activity induces brain functional changes and neuroplasticity, leading to an improvement of cognitive functions, such as learning and memory in the offspring. This study investigated the effects of voluntary maternal physical activity on the gene expression of the neurotrophic factors (NTFs): BDNF, NTF4, NTRK2, IGF-1 and IGF-1r in the different areas of mother's brain, placenta and foetus brain of rats. Female Wistar rats (n = 15) were individually housed in voluntary physical activity cages, containing a running wheel, for 4 weeks (period of adaptation) before gestation. Rats were classified as inactive (I, n = 6); active (A, n = 4) and very active (VA, n = 5) according to daily distance spontaneously travelled. During gestation, the dams continued to have access to the running wheel. At the 20th day of gestation, gene expression of NTFs was analysed in different areas of mother's brain (cerebellum, hypothalamus, hippocampus and cortex), placenta and the offspring's brain. NTFs gene expression was evaluated using quantitative PCR. Very active mothers showed upregulation of IGF-1 mRNA in the cerebellum (36.8%) and NTF4 mRNA expression in the placenta (24.3%). In the cortex, there was a tendency of up-regulation of NTRK2 mRNA (p = 0.06) in the A and VA groups when compared to I group. There were no noticeable changes in the gene expression of NTFs in the offspring's brain. Our findings suggest the existence of a developmental plasticity induced by maternal physical activity in specific areas of the brain and placenta representing the first investment for offspring during development.
Collapse
|
27
|
Keshavarz E, Motevasselian M, Amirnazeri B, Bahramzadeh S, Mohammadkhani H, Mehrjardi Z, Razzaz M, Bakhtiyari M. Gestational age-specific reference values of placental thickness in normal pregnant women. Women Health 2019; 59:718-729. [PMID: 30786837 DOI: 10.1080/03630242.2018.1553816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The aim of this study was to determine the reference values of the placental thickness in 400 normal pregnant women during weeks 16-41 of gestation who were referred to the Mahdiyeh hospital during January 2014-February 2015. The placental thickness at the junction of the umbilical artery was measured using ultrasonography. Multivariable linear regression was used to model the mean placental thickness and assess associations with measured covariates, including gestational age (GA). Centiles for placental thickness distribution were estimated according to the modulus exponential-normal model. The mean and standard deviation of the age of the participants was 31.4 ± 5.7 years. The Pearson correlation coefficient indicated a very strong positive linear correlation between GA and placental thickness (p <.001; r = 0.93). A nonlinear increase of placental thickness with estimated fetal weights was observed. GA-specific placental thicknesses for the 2.5th, 5th, 10th, 25th, 50th, 75th, 90th, 95th, and 97.5th percentiles of placental thickness were calculated. By making use of the provided nomogram in this study, neonatal outcomes associated with the placental thickness, such as Hemoglobin Bart's disease, can be diagnosed early to improve maternal and newborn health.
Collapse
Affiliation(s)
- E Keshavarz
- a Department of Radiology , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - M Motevasselian
- b Department of Gynecology & Obstetric , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - B Amirnazeri
- b Department of Gynecology & Obstetric , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - S Bahramzadeh
- b Department of Gynecology & Obstetric , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - H Mohammadkhani
- b Department of Gynecology & Obstetric , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Z Mehrjardi
- b Department of Gynecology & Obstetric , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - M Razzaz
- c Department of Radiology , Kerman University of Medical Sciences , Kerman , Iran
| | - M Bakhtiyari
- d Non-Communicable Diseases Research Center , Alborz University of Medical Sciences , Karaj , Iran.,e Department of Community Medicine , Alborz University of Medical Sciences , Karaj , Iran
| |
Collapse
|
28
|
Morano D, Rossi S, Lapucci C, Pittalis MC, Farina A. Cell-Free DNA (cfDNA) Fetal Fraction in Early- and Late-Onset Fetal Growth Restriction. Mol Diagn Ther 2019; 22:613-619. [PMID: 30056492 DOI: 10.1007/s40291-018-0353-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Our objective was to retrospectively evaluate whether the levels of cell-free DNA (cfDNA) fetal fraction differed in the first trimester of pregnancies between controls and those who subsequently developed early- or late-onset fetal growth restriction (FGR). METHODS This was a case-control study conducted between May 2015 and May 2018 in 231 low-risk women who had received first trimester screening for major fetal aneuploidies (Panorama, Natera, San Carlos, CA, USA). Early- and late-onset FGR developed in 5 and 16 women, respectively, according to Delphi criteria. Multiples of median (MoM) were used to evaluate the differences in cfDNA fetal fraction between cases and controls. cfDNA fetal fraction was adjusted for gestational age (from 10 + 0 to 13 + 6 gestational weeks) and maternal weight (43-96 kg). RESULTS The median cfDNA fetal fractions for controls and early- and late-onset FGR were 1.00 (interquartile range [IQR] 0.89-1.12), 0.69 (IQR 0.44-0.84) and 0.93 (IQR 0.83-1.03) MoM, respectively. Statistically lower cfDNA fetal fraction MoM values were observed only in patients with early-onset FGR (Kruskal-Wallis test with Dunn post hoc test). In a 1:35 ratio (one case of early-onset FGR: 35 controls), the mean observed rank of 2.00 ± 2.23 in the cases was significantly lower than the expected 18.97 ± 10.17 (p < 0.001). CONCLUSIONS Low-risk pregnancies that developed early-onset FGR had lower cfDNA fetal fractions than did the matched controls. This result is consistent with the placental dysfunction typical of early-onset FGR. For possible clinical use, the cfDNA fetal fraction would yield a better predictive value if adjusted for maternal weight, since maternal weight affects both cfDNA fetal fraction and the occurrence of FGR.
Collapse
Affiliation(s)
- Danila Morano
- Azienda Ospedaliero-Universitaria S. Anna, Cona, Ferrara, Italy
| | - Stefania Rossi
- Division of Obstetrics and Prenatal Medicine, Department of Medicine and Surgery (DIMEC), Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy.,Poliambulatorio SaluSanGiorgio, San Giorgio di Piano, Bologna, Italy
| | | | - Maria Carla Pittalis
- Division of Obstetrics and Prenatal Medicine, Department of Medicine and Surgery (DIMEC), Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Antonio Farina
- Division of Obstetrics and Prenatal Medicine, Department of Medicine and Surgery (DIMEC), Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy.
| |
Collapse
|
29
|
van Donge T, Evers K, Koch G, van den Anker J, Pfister M. Clinical Pharmacology and Pharmacometrics to Better Understand Physiological Changes During Pregnancy and Neonatal Life. Handb Exp Pharmacol 2019; 261:325-337. [PMID: 30968215 DOI: 10.1007/164_2019_210] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Pregnant women, fetuses, and newborns are particularly vulnerable patient populations. During pregnancy, the body is subject to physiological changes that influence the pharmacokinetics and pharmacodynamics of drugs. Inappropriate dosing in pregnant women can result in sub-therapeutic or toxic effects, putting not only the pregnant woman but also her fetus at risk. During neonatal life, maturation processes also affect pharmacokinetics and pharmacodynamics of drugs. Inappropriate dosing in newborns leads not only to short-term complications but can also have a negative impact on the long-term development of infants and children. For these reasons, it is crucial to characterize physiological changes in pregnant women, describe placental transfer kinetics of drugs, and describe physiological changes related to the transition from intrauterine to extrauterine life and maturation processes in preterm and term neonates. Quantitative pharmacological approaches such as pharmacometric and physiologically-based modeling and model-based simulations can be useful to better understand and predict such physiological changes and their effects on drug exposure and response. This review article (1) gives an overview of physiological changes in pregnant women, their fetuses, and (pre)term neonates, (2) presents case studies to illustrate applications of new modeling and simulation approaches, and (3) discusses challenges and opportunities in optimizing and personalizing treatments during pregnancy and neonatal life.
Collapse
Affiliation(s)
- Tamara van Donge
- Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland.
| | - Katrina Evers
- Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland
| | - Gilbert Koch
- Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland
| | - John van den Anker
- Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland.,Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands.,Division of Clinical Pharmacology, Children's National Health System, Washington, DC, USA
| | - Marc Pfister
- Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland.,Certara LP, Princeton, NJ, USA
| |
Collapse
|
30
|
Tossetta G, Fantone S, Giannubilo SR, Marinelli Busilacchi E, Ciavattini A, Castellucci M, Di Simone N, Mattioli-Belmonte M, Marzioni D. Pre-eclampsia onset and SPARC: A possible involvement in placenta development. J Cell Physiol 2018; 234:6091-6098. [PMID: 30426491 DOI: 10.1002/jcp.27344] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/10/2018] [Indexed: 12/22/2022]
Abstract
Pre-eclampsia (PE) is a multisystem disorder commonly diagnosed in the latter half of pregnancy and it is a leading cause of intrauterine fetal growth retardation (IUGR). The aim of this study was to investigate the localization and the role of SPARC, secreted protein acidic, and rich in cysteine, in PE and PE-IUGR placentas in comparison with normal placentas. SPARC was mainly expressed in the villous and extravillous cytotrophoblastic cells in first trimester, whereas in PE, PE-IUGR and at term placentas, SPARC immunostaining was visible in both cytotrophoblastic cells and syncytiotrophoblast. SPARC expression significantly decreased in normal placenta from first to third trimester and a further significant reduction was demonstrated in PE and PE-IUGR. The latter downregulation of SPARC depends on hypoxic condition as shown by in vitro models. In conclusion, SPARC can play a pivotal role in PE and PE-IUGR onset and it should be considered as a key molecule for future investigations in such pathologies.
Collapse
Affiliation(s)
- Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Sonia Fantone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Stefano R Giannubilo
- Department of Clinical Science, Università Politecnica delle Marche, Ancona, Italy
| | | | - Andrea Ciavattini
- Department of Clinical Science, Università Politecnica delle Marche, Ancona, Italy
| | - Mario Castellucci
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Nicoletta Di Simone
- Department of Woman and Child Health, A. Gemelli Hospital, Università Cattolica Del Sacro Cuore of Rome, Rome, Italy
| | - Monica Mattioli-Belmonte
- Department of Molecular and Clinical Sciences, Università Politenica delle Marche, Ancona, Italy
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
31
|
Konwar C, Price EM, Wang LQ, Wilson SL, Terry J, Robinson WP. DNA methylation profiling of acute chorioamnionitis-associated placentas and fetal membranes: insights into epigenetic variation in spontaneous preterm births. Epigenetics Chromatin 2018; 11:63. [PMID: 30373633 PMCID: PMC6205793 DOI: 10.1186/s13072-018-0234-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/20/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Placental inflammation, often presenting as acute chorioamnionitis (aCA), is commonly associated with preterm birth. Preterm birth can have both immediate and long-term adverse effects on the health of the baby. Developing biomarkers of inflammation in the placenta can help to understand its effects and potentially lead to new approaches for rapid prenatal diagnosis of aCA. We aimed to characterize epigenetic variation associated with aCA in placenta (chorionic villi) and fetal membranes (chorion and amnion) to better understand how aCA may impact processes that lead to preterm birth. This study lays the groundwork for development of novel biomarkers for aCA. METHODS Samples from 44 preterm placentas (chorionic villi) as well as matched chorion and amnion for 16 of these cases were collected for this study. These samples were profiled using the Illumina Infinium HumanMethylation850 BeadChip to measure DNA methylation (DNAm) at 866,895 CpGs across the genome. An additional 78 placental samples were utilized to independently validate the array findings by pyrosequencing. RESULTS Using a false discovery rate of < 0.15 and average group difference in DNAm of > 0.05, 66 differentially methylated (DM) CpG sites were identified between aCA cases and non-aCA cases in chorionic villi. For the majority of these 66 DM CpGs, the DNAm profile of the aCA cases as compared to the non-aCA cases trended in the direction of the blood cell DNAm. Interestingly, neutrophil-specific DNAm signatures, but not those associated with other immune cell types, were capable of separating aCA cases from the non-aCA cases. CONCLUSIONS Our results suggest that aCA-associated placentas showed altered DNAm signatures that were not observed in the absence of aCA. This DNAm profile is consistent with the activation of the innate immune response in the placenta and/or reflect increase in neutrophils as a response to inflammation.
Collapse
Affiliation(s)
- Chaini Konwar
- BC Children’s Hospital Research Institute, 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1 Canada
| | - E. Magda Price
- BC Children’s Hospital Research Institute, 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1 Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3N1 Canada
| | - Li Qing Wang
- BC Children’s Hospital Research Institute, 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada
- University of British Columbia, Vancouver, BC V6H 3N1 Canada
| | - Samantha L. Wilson
- BC Children’s Hospital Research Institute, 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1 Canada
| | - Jefferson Terry
- BC Children’s Hospital Research Institute, 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada
- Department of Pathology, BC Children’s Hospital, Vancouver, BC V6H 3N1 Canada
| | - Wendy P. Robinson
- BC Children’s Hospital Research Institute, 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1 Canada
| |
Collapse
|
32
|
Gilman-Sachs A, Dambaeva S, Salazar Garcia MD, Hussein Y, Kwak-Kim J, Beaman K. Inflammation induced preterm labor and birth. J Reprod Immunol 2018; 129:53-58. [PMID: 30025845 DOI: 10.1016/j.jri.2018.06.029] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 06/25/2018] [Indexed: 01/23/2023]
Abstract
Preterm birth which occurs before 37 weeks gestation is one of the most common obstetrical complication in humans. After many studies, it appears that "not one answer fits all" regarding the risk factors, causes and the treatments for this syndrome. However, it is becoming more evident that one of the major risk factors is inflammation and/or infection in the fetoplacental unit. In animal models (usually consisting of mice injected with lipopolysaccharide at 14 days of gestation), IL-22 and IL-6 have been identified as factors related to preterm birth. There are some clinical tests available to determine the risk for preterm labor and delivery, which can be identified before, during early, or at mid-gestation. However, treatment of preterm birth with antibiotics so far has not been "curable" and studies using anti-inflammatory treatments are not readily available. More studies regarding causes and treatments for preterm labor and delivery in humans are necessary to prevent neonatal deaths and/or developmental abnormalities associated with this common syndrome.
Collapse
Affiliation(s)
- Alice Gilman-Sachs
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, United States.
| | - Svetlana Dambaeva
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, United States
| | - Maria D Salazar Garcia
- Reproductive Medicine and Immunology, Department of Obstetrics and Gynecology, Chicago Medicine School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, United States
| | - Youssef Hussein
- Reproductive Medicine and Immunology, Department of Obstetrics and Gynecology, Chicago Medicine School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, United States
| | - Joanne Kwak-Kim
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, United States; Reproductive Medicine and Immunology, Department of Obstetrics and Gynecology, Chicago Medicine School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, United States
| | - Kenneth Beaman
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, United States
| |
Collapse
|