1
|
Afrose D, Alfonso-Sánchez S, McClements L. Targeting oxidative stress in preeclampsia. Hypertens Pregnancy 2025; 44:2445556. [PMID: 39726411 DOI: 10.1080/10641955.2024.2445556] [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: 09/21/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Preeclampsia is a complex condition characterized by elevated blood pressure and organ damage involving kidneys or liver, resulting in significant morbidity and mortality for both the mother and the fetus. Increasing evidence suggests that oxidative stress, often caused by mitochondrial dysfunction within fetal trophoblast cells may play a major role in the development and progression of preeclampsia. Oxidative stress occurs as a result of an imbalance between the production of reactive oxygen species (ROS) and the capacity of antioxidant defenses, which can lead to placental cellular damage and endothelial cell dysfunction. Targeting oxidative stress appears to be a promising therapeutic approach that has the potential to improve both short- and long-term maternal and fetal outcomes, thus reducing the global burden of preeclampsia. The purpose of this review is to provide a comprehensive account of the mechanisms of oxidative stress in preeclampsia. Furthermore, it also examines potential interventions for reducing oxidative stress in preeclampsia, including natural antioxidant supplements, lifestyle modifications, mitochondrial targeting antioxidants, and pharmacological agents.A better understanding of the mechanism of action of proposed therapeutic strategies targeting oxidative stress is essential for the identification of companion biomarkers and personalized medicine approaches for the development of effective treatments of preeclampsia.
Collapse
Affiliation(s)
- Dinara Afrose
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Sofía Alfonso-Sánchez
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
2
|
Vincent KMM, Garner T, Stevens A, Cottrell EC, Myers JE, Higgins LE. Assessing Placental Dysfunction Subtypes in Pregnancies With a Low PlGF Centile. Hypertension 2025. [PMID: 40396254 DOI: 10.1161/hypertensionaha.124.24440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/29/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND A low circulating placental growth factor (PlGF) concentration is considered a marker of placental dysfunction, the predominant cause of preeclampsia and fetal growth restriction. Besides iatrogenic delivery, there are no effective treatments, potentially due to its heterogeneity. We hypothesize that >1 subtype of placental dysfunction exists in pregnancies with a low circulating PlGF. METHODS Matched placental villous and maternal plasma samples (low PlGF <5th, n=19; normal PlGF ≥5th centile, n=20) were collected from uncomplicated pregnancies and those complicated by 1 or a combination of preeclampsia, fetal growth restriction, or chronic hypertension. Transcriptomic and metabolomic data sets were obtained from villous samples and used to perform cluster-of-cluster analysis. Clinical outcomes were compared between clusters, as well as subsequent pathway analysis. RESULTS Multiomic cluster-of-cluster analysis resulted in 3 molecular clusters. Cluster 1 predominantly consisted of those with a low PlGF (9/10); PlGF results in cluster 2 varied (low, n=8/20; normal, n=12/20), while cluster 3 mainly comprised of those with a normal PlGF result (n=7/9). Birthweight was significantly lower in cluster 1, as well as an increased incidence of early-onset preeclampsia. However, pathways commonly associated with placental dysfunction were only identified in cluster 2. CONCLUSIONS Multiomic analysis revealed 2 potential subtypes of placental dysfunction associated with a low circulating PlGF. These results support previous studies suggesting the existence of preeclampsia subtypes. Clinical outcome comparisons indicate cluster 1 as the severe subtype; however, pathway analysis contradicted this. Improved understanding of subtype etiology could enable a more personalized therapeutic approach for pregnancies complicated by placental dysfunction.
Collapse
Affiliation(s)
- Kirsty M M Vincent
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, United Kingdom (K.M.M.V., T.G., A.S., E.C.C., J.E.M., L.E.H.)
| | - Terence Garner
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, United Kingdom (K.M.M.V., T.G., A.S., E.C.C., J.E.M., L.E.H.)
| | - Adam Stevens
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, United Kingdom (K.M.M.V., T.G., A.S., E.C.C., J.E.M., L.E.H.)
| | - Elizabeth C Cottrell
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, United Kingdom (K.M.M.V., T.G., A.S., E.C.C., J.E.M., L.E.H.)
| | - Jenny E Myers
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, United Kingdom (K.M.M.V., T.G., A.S., E.C.C., J.E.M., L.E.H.)
- Manchester University Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, United Kingdom (J.E.M., L.E.H.)
| | - Lucy E Higgins
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, United Kingdom (K.M.M.V., T.G., A.S., E.C.C., J.E.M., L.E.H.)
- Manchester University Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, United Kingdom (J.E.M., L.E.H.)
| |
Collapse
|
3
|
Huang T, Lu F. Genetically predicted circulating concentrations of micronutrients and risk of hypertensive disorders of pregnancy: a Mendelian randomization study. Arch Gynecol Obstet 2024; 310:1019-1025. [PMID: 38194093 DOI: 10.1007/s00404-023-07331-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 12/01/2023] [Indexed: 01/10/2024]
Abstract
PURPOSE Epidemiological studies examining the association between circulating micronutrients and the risk of hypertensive disorders during pregnancy (HDP) have produced inconsistent results. Therefore, we conducted a Mendelian randomization (MR) analysis to evaluate the potential causal relationship between micronutrients and HDP. METHODS Nine micronutrients (beta-carotene, vitamin B6, vitamin B12, calcium, zinc, selenium, copper, folate, and phosphorus) were selected as the exposure factors. Summary data for gestational hypertension (14,727 cases and 196,143 controls) and preeclampsia/eclampsia (7212 cases and 174,266 controls) were extracted from the FinnGen consortium. The MR analysis employed the inverse variance weighted method and conducted a range of sensitivity analyses. RESULTS The inverse variance weighted method indicated no significant causal relationship between nine genetically predicted micronutrient concentrations and gestational hypertension, as well as preeclampsia/eclampsia. Sensitivity analyses suggested the absence of pleiotropy. CONCLUSION There is no strong evidence to support the causation between circulating micronutrients and hypertensive disorder during pregnancy.
Collapse
Affiliation(s)
- Ting Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Fan Lu
- Department of Emergency, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
4
|
Allotey J, Archer L, Coomar D, Snell KI, Smuk M, Oakey L, Haqnawaz S, Betrán AP, Chappell LC, Ganzevoort W, Gordijn S, Khalil A, Mol BW, Morris RK, Myers J, Papageorghiou AT, Thilaganathan B, Da Silva Costa F, Facchinetti F, Coomarasamy A, Ohkuchi A, Eskild A, Arenas Ramírez J, Galindo A, Herraiz I, Prefumo F, Saito S, Sletner L, Cecatti JG, Gabbay-Benziv R, Goffinet F, Baschat AA, Souza RT, Mone F, Farrar D, Heinonen S, Salvesen KÅ, Smits LJ, Bhattacharya S, Nagata C, Takeda S, van Gelder MM, Anggraini D, Yeo S, West J, Zamora J, Mistry H, Riley RD, Thangaratinam S. Development and validation of prediction models for fetal growth restriction and birthweight: an individual participant data meta-analysis. Health Technol Assess 2024; 28:1-119. [PMID: 39252507 PMCID: PMC11404361 DOI: 10.3310/dabw4814] [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] [Indexed: 09/11/2024] Open
Abstract
Background Fetal growth restriction is associated with perinatal morbidity and mortality. Early identification of women having at-risk fetuses can reduce perinatal adverse outcomes. Objectives To assess the predictive performance of existing models predicting fetal growth restriction and birthweight, and if needed, to develop and validate new multivariable models using individual participant data. Design Individual participant data meta-analyses of cohorts in International Prediction of Pregnancy Complications network, decision curve analysis and health economics analysis. Participants Pregnant women at booking. External validation of existing models (9 cohorts, 441,415 pregnancies); International Prediction of Pregnancy Complications model development and validation (4 cohorts, 237,228 pregnancies). Predictors Maternal clinical characteristics, biochemical and ultrasound markers. Primary outcomes fetal growth restriction defined as birthweight <10th centile adjusted for gestational age and with stillbirth, neonatal death or delivery before 32 weeks' gestation birthweight. Analysis First, we externally validated existing models using individual participant data meta-analysis. If needed, we developed and validated new International Prediction of Pregnancy Complications models using random-intercept regression models with backward elimination for variable selection and undertook internal-external cross-validation. We estimated the study-specific performance (c-statistic, calibration slope, calibration-in-the-large) for each model and pooled using random-effects meta-analysis. Heterogeneity was quantified using τ2 and 95% prediction intervals. We assessed the clinical utility of the fetal growth restriction model using decision curve analysis, and health economics analysis based on National Institute for Health and Care Excellence 2008 model. Results Of the 119 published models, one birthweight model (Poon) could be validated. None reported fetal growth restriction using our definition. Across all cohorts, the Poon model had good summary calibration slope of 0.93 (95% confidence interval 0.90 to 0.96) with slight overfitting, and underpredicted birthweight by 90.4 g on average (95% confidence interval 37.9 g to 142.9 g). The newly developed International Prediction of Pregnancy Complications-fetal growth restriction model included maternal age, height, parity, smoking status, ethnicity, and any history of hypertension, pre-eclampsia, previous stillbirth or small for gestational age baby and gestational age at delivery. This allowed predictions conditional on a range of assumed gestational ages at delivery. The pooled apparent c-statistic and calibration were 0.96 (95% confidence interval 0.51 to 1.0), and 0.95 (95% confidence interval 0.67 to 1.23), respectively. The model showed positive net benefit for predicted probability thresholds between 1% and 90%. In addition to the predictors in the International Prediction of Pregnancy Complications-fetal growth restriction model, the International Prediction of Pregnancy Complications-birthweight model included maternal weight, history of diabetes and mode of conception. Average calibration slope across cohorts in the internal-external cross-validation was 1.00 (95% confidence interval 0.78 to 1.23) with no evidence of overfitting. Birthweight was underestimated by 9.7 g on average (95% confidence interval -154.3 g to 173.8 g). Limitations We could not externally validate most of the published models due to variations in the definitions of outcomes. Internal-external cross-validation of our International Prediction of Pregnancy Complications-fetal growth restriction model was limited by the paucity of events in the included cohorts. The economic evaluation using the published National Institute for Health and Care Excellence 2008 model may not reflect current practice, and full economic evaluation was not possible due to paucity of data. Future work International Prediction of Pregnancy Complications models' performance needs to be assessed in routine practice, and their impact on decision-making and clinical outcomes needs evaluation. Conclusion The International Prediction of Pregnancy Complications-fetal growth restriction and International Prediction of Pregnancy Complications-birthweight models accurately predict fetal growth restriction and birthweight for various assumed gestational ages at delivery. These can be used to stratify the risk status at booking, plan monitoring and management. Study registration This study is registered as PROSPERO CRD42019135045. Funding This award was funded by the National Institute for Health and Care Research (NIHR) Health Technology Assessment programme (NIHR award ref: 17/148/07) and is published in full in Health Technology Assessment; Vol. 28, No. 14. See the NIHR Funding and Awards website for further award information.
Collapse
Affiliation(s)
- John Allotey
- WHO Collaborating Centre for Global Women's Health, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Lucinda Archer
- Centre for Prognosis Research, School of Medicine, Keele University, Keele, UK
| | - Dyuti Coomar
- WHO Collaborating Centre for Global Women's Health, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Kym Ie Snell
- Centre for Prognosis Research, School of Medicine, Keele University, Keele, UK
| | - Melanie Smuk
- Blizard Institute, Centre for Genomics and Child Health, Queen Mary University of London, London, UK
| | - Lucy Oakey
- WHO Collaborating Centre for Global Women's Health, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Sadia Haqnawaz
- The Hildas, Dame Hilda Lloyd Network, WHO Collaborating Centre for Global Women's Health, University of Birmingham, Birmingham, UK
| | - Ana Pilar Betrán
- Department of Reproductive and Health Research, World Health Organization, Geneva, Switzerland
| | - Lucy C Chappell
- Department of Women and Children's Health, School of Life Course Sciences, King's College London, London, UK
| | - Wessel Ganzevoort
- Department of Obstetrics, Amsterdam UMC University of Amsterdam, Amsterdam, the Netherlands
| | - Sanne Gordijn
- Faculty of Medical Sciences, University Medical Center Groningen, Groningen, the Netherlands
| | - Asma Khalil
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust and Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Ben W Mol
- Department of Obstetrics and Gynaecology, Monash University, Monash Medical Centre, Clayton, Victoria, Australia
- Aberdeen Centre for Women's Health Research, Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - Rachel K Morris
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Jenny Myers
- Maternal and Fetal Health Research Centre, Manchester Academic Health Science Centre, University of Manchester, Central Manchester NHS Trust, Manchester, UK
| | - Aris T Papageorghiou
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust and Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Basky Thilaganathan
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust and Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
- Tommy's National Centre for Maternity Improvement, Royal College of Obstetrics and Gynaecology, London, UK
| | - Fabricio Da Silva Costa
- Maternal Fetal Medicine Unit, Gold Coast University Hospital and School of Medicine, Griffith University, Gold Coast, Queensland, Australia
| | - Fabio Facchinetti
- Mother-Infant Department, University of Modena and Reggio Emilia, Emilia-Romagna, Italy
| | - Arri Coomarasamy
- WHO Collaborating Centre for Global Women's Health, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Akihide Ohkuchi
- Department of Obstetrics and Gynecology, Jichi Medical University School of Medicine, Shimotsuke-shi, Tochigi, Japan
| | - Anne Eskild
- Akershus University Hospital, University of Oslo, Oslo, Norway
| | | | - Alberto Galindo
- Fetal Medicine Unit, Maternal and Child Health and Development Network (SAMID), Department of Obstetrics and Gynaecology, Hospital Universitario, Instituto de Investigación Hospital, Universidad Complutense de Madrid, Madrid, Spain
| | - Ignacio Herraiz
- Department of Obstetrics and Gynaecology, Hospital Universitario, Madrid, Spain
| | - Federico Prefumo
- Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Shigeru Saito
- Department Obstetrics and Gynecology, University of Toyama, Toyama, Japan
| | - Line Sletner
- Deptartment of Pediatric and Adolescents Medicine, Akershus University Hospital, Sykehusveien, Norway
| | - Jose Guilherme Cecatti
- Obstetric Unit, Department of Obstetrics and Gynecology, University of Campinas, Campinas, Sao Paulo, Brazil
| | - Rinat Gabbay-Benziv
- Maternal Fetal Medicine Unit, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center Hadera, Affiliated to the Ruth and Bruce Rappaport School of Medicine, Technion, Haifa, Israel
| | - Francois Goffinet
- Maternité Port-Royal, AP-HP, APHP, Centre-Université de Paris, FHU PREMA, Paris, France
- Université de Paris, INSERM U1153, Equipe de recherche en Epidémiologie Obstétricale, Périnatale et Pédiatrique (EPOPé), Centre de Recherche Epidémiologie et Biostatistique Sorbonne Paris Cité (CRESS), Paris, France
| | - Ahmet A Baschat
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, MD, USA
| | - Renato T Souza
- Obstetric Unit, Department of Obstetrics and Gynecology, University of Campinas, Campinas, Sao Paulo, Brazil
| | - Fionnuala Mone
- Centre for Public Health, Queen's University, Belfast, UK
| | - Diane Farrar
- Bradford Institute for Health Research, Bradford, UK
| | - Seppo Heinonen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kjell Å Salvesen
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Luc Jm Smits
- Care and Public Health Research Institute, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Sohinee Bhattacharya
- Aberdeen Centre for Women's Health Research, Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - Chie Nagata
- Center for Postgraduate Education and Training, National Center for Child Health and Development, Tokyo, Japan
| | - Satoru Takeda
- Department of Obstetrics and Gynecology, Juntendo University, Tokyo, Japan
| | - Marleen Mhj van Gelder
- Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Dewi Anggraini
- Faculty of Mathematics and Natural Sciences, Lambung Mangkurat University, South Kalimantan, Indonesia
| | - SeonAe Yeo
- University of North Carolina at Chapel Hill, School of Nursing, NC, USA
| | - Jane West
- Bradford Institute for Health Research, Bradford, UK
| | - Javier Zamora
- WHO Collaborating Centre for Global Women's Health, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, Spain
| | - Hema Mistry
- Warwick Medical School, University of Warwick, Warwick, UK
| | - Richard D Riley
- Centre for Prognosis Research, School of Medicine, Keele University, Keele, UK
| | - Shakila Thangaratinam
- WHO Collaborating Centre for Global Women's Health, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
5
|
Sen S, Cherkerzian S, Herlihy M, Hacker MR, McElrath TF, Cantonwine DE, Fichorova R, Oken E, Meydani SN. Supplementation with antioxidant micronutrients in pregnant women with obesity: a randomized controlled trial. Int J Obes (Lond) 2024; 48:796-807. [PMID: 38396126 DOI: 10.1038/s41366-024-01472-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/03/2024] [Accepted: 01/15/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND/OBJECTIVE Obesity increases maternal morbidity and adversely affects child health. Maternal inflammation may play a role in adverse outcomes. The objective of this study was to determine whether providing a higher dose of antioxidant micronutrients to pregnant women with obesity would raise concentrations of key antioxidant vitamins and impact inflammation and oxidative stress during pregnancy. SUBJECTS/METHODS This was a double-blind, randomized controlled trial. We recruited pregnant women with a body mass index (BMI) ≥ 30 kg/m2 at their initial prenatal visit ( < 13 weeks gestation) and collected blood and urine samples at baseline, 24-28 weeks, and 32-36 weeks to measure micronutrient concentrations (vitamin C, E, B6 and folate), markers of inflammation (C-reactive protein, interleukin-6, 8, and 1β) and oxidative stress (8-epi-PGF2α and malondialdehyde). We collected maternal and infant health data from enrollment to delivery as secondary outcomes. We enrolled 128 participants (64 in each arm), and 98 (49 in each arm) completed follow-up through delivery. INTERVENTION Both groups received a standard prenatal vitamin containing the recommended daily allowance of micronutrients in pregnancy. In addition, the intervention group received a supplement with 90 mg vitamin C, 30 αTU vitamin E, 18 mg vitamin B6, and 800 μg folic acid, and the control group received a placebo. RESULTS The intervention group had higher vit B6 (log transformed (ln), β 24-28 weeks: 0.76 nmol/L (95% CI: 0.40, 1.12); β 32-36 weeks: 0.52 nmol/L (95% CI: 0.17, 0.88)) than the control group. Vitamins C, E, erythrocyte RBC folate concentrations did not differ by randomization group. The intervention did not impact biomarkers of inflammation or oxidative stress. There were no differences in maternal or neonatal clinical outcomes by randomization group. CONCLUSIONS Higher concentrations of antioxidant vitamins during pregnancy increased specific micronutrients and did not impact maternal inflammation and oxidative stress, which may be related to dosing or type of supplementation provided. CLINICAL TRIAL REGISTRATION Clinical Trial Identification Number: NCT02802566; URL of the Registration Site: www. CLINICALTRIALS gov .
Collapse
Affiliation(s)
- Sarbattama Sen
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- JM USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Sara Cherkerzian
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Mary Herlihy
- Harvard Medical School, Boston, MA, USA
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Michele R Hacker
- Harvard Medical School, Boston, MA, USA
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Thomas F McElrath
- Harvard Medical School, Boston, MA, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Boston, MA, USA
| | - David E Cantonwine
- Harvard Medical School, Boston, MA, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Boston, MA, USA
| | - Raina Fichorova
- Harvard Medical School, Boston, MA, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Boston, MA, USA
| | - Emily Oken
- Harvard Medical School, Boston, MA, USA
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Simin N Meydani
- JM USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| |
Collapse
|
6
|
Movaghar R, Abbasalizadeh S, Vazifekhah S, Farshbaf-Khalili A, Shahnazi M. The effects of synbiotic supplementation on blood pressure and other maternal outcomes in pregnant mothers with mild preeclampsia: a triple-blinded randomized controlled trial. BMC Womens Health 2024; 24:80. [PMID: 38297273 PMCID: PMC10829212 DOI: 10.1186/s12905-024-02922-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 01/21/2024] [Indexed: 02/02/2024] Open
Abstract
INTRODUCTION Preeclampsia affects a significant percentage of pregnancies which is a leading cause of premature birth. Probiotics have the potential to affect inflammatory factors, and oxidative stress, which are linked to the development of preeclampsia. The study aimed to compare the effect of synbiotic and placebo on blood pressure and pregnancy duration as primary outcomes, and other pregnancy outcomes. METHODS This study comprised 128 pregnant women with mild preeclampsia and gestational ages exceeding 24 weeks who were referred to the high-risk pregnancy clinic. It was a randomized, controlled, phase III, triple-blinded clinical experiment. The intervention and control groups were distributed to the participants at random. Intervention group received one oral synbiotic capsule, and control group received placebo daily until delivery. Based on gestational age at the time of diagnosis, preeclampsia was stratificated as early (< 34 weeks) or late (≥ 34 weeks). Data obtained from questionnaires, and biochemical serum factors were analyzed using SPSS software version 23 software. RESULTS With the exception of the history of taking vitamin D3, there were no statistically significant variations in socio-demographic variables between the research groups. After the intervention, the means of systolic blood pressure (adjusted mean difference: -13.54, 95% CI: -5.01 to -22.07), and diastolic blood pressure (adjusted mean difference: -10.30, 95% CI: -4.70 to -15.90) were significantly lower in the synbiotic-supplemented group than in the placebo group. Compared to the placebo group, the incidence of severe PE (p < 0.001), proteinuria (p = 0.044), and mean serum creatinine level (p = 0.005) significantly declined in the synbiotic-supplemented group after the intervention. However, our analysis found no significant association for other outcomes. CONCLUSION Based on our results, synbiotic had beneficial effects on some pregnancy outcomes. Further studies with larger samples are needed to verify the advantages of synbiotic supplementation for high-risk pregnancies, particularly with regards to higher doses, and longer intervention periods. TRIAL REGISTRATION IRCT20110606006709N20.
Collapse
Affiliation(s)
- Rouhina Movaghar
- Department of Midwifery, Faculty of Midwifery, Mahabad Branch Azad University, Mahabad, Iran
| | - Shamci Abbasalizadeh
- Tabriz University of Medical Sciences, Women's Health Research Center, Tabriz, Iran
| | | | - Azizeh Farshbaf-Khalili
- Physical Medicine and Rehabilitation Research Centre, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, IR, Iran.
| | - Mahnaz Shahnazi
- Faculty of Nursing and Midwifery, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
7
|
Salem AA, Taha DA, Nasr AA, El-Sagheer M, Daghash W, Taghian RA. Effect of vitamin E, D-limonene, and their combination on nulliparous rabbit reproductive performance. Anim Reprod Sci 2023; 259:107378. [PMID: 37989002 DOI: 10.1016/j.anireprosci.2023.107378] [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: 09/08/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023]
Abstract
Oxidative status is important in reproductive performance and using two natural antioxidants is more beneficial than one in nulliparous rabbits. The goal, effect of vitamin E (VitE), D-limonene (DL), and VitE+DL on maternal LBW (MLBW), conception (CR), pregnancy (PR), and kindling rates (KR), gestation length (GL), total litter size at birth (TLSB) and weaning (TLSW), live kits at birth (LKB) and weaning (LKW), dead kits at birth (DKB) and weaning (DKW), kits mortality rate at weaning (KMRW), Kit weight at birth (KWB) and weaning (KWW), total kit weight at birth (TKWB) and weaning (TKWW), and concentrations of progesterone (P4) and Malondialdehyde (MDA), during first two pregnancies. A total of 24 healthy female WNZ rabbits were randomly selected and assigned into four groups (6/each). Control (animals injected with 1.0 mL propylene glycol), VitE (60 mg IM injection/animal, 2X/week pre-mating and 3X post-mating until mid-pregnancy, DL (20 mg IM injection/animal, 2X/week pre-mating and 1X at mating, and VitE+DL (IM injection/animal with the same doses and times applied in VitE and DL groups. All animals were treated during 1st pregnancy only. The results confirmed that animals treated with VitE+DL gained significant maternal LBW in 1st pregnancy, reduced dead kit number at birth and kit mortality rate at weaning, increased live kits and total kit weight at birth and weaning in the two pregnancies, and also increased significantly kit weight at birth and weaning in the treatments than controls in the two pregnancies, and DL was greater in 1st pregnancy. Progesterone concentrations in mid-pregnancy rose significantly in VitE+DL during 1st pregnancy and DL in 2nd pregnancy. Malondialdehyde concentrations dropped significantly in VitE and VitE+DL in mid-pregnancy in the two pregnancies. Eventually, the integration of VitE and DL displayed their unique properties for improving productive and reproductive performance in nulliparous rabbits.
Collapse
Affiliation(s)
- Anas A Salem
- Animal Production Department, Fac. of Agriculture, Assiut University, Assiut, Egypt.
| | - Doaa A Taha
- Animal Production Department, Fac. of Agriculture, Assiut University, Assiut, Egypt.
| | - Asmaa Ae Nasr
- Pathology Department, Institute of Veterinary Health, Assiut, Egypt.
| | - Mohamed El-Sagheer
- Poultry Production Department, Fac. of Agriculture, Assiut University, Egypt
| | - Wael Daghash
- Animal Production Department, Fac. of Agriculture, Assiut University, Assiut, Egypt
| | - Raghda A Taghian
- Animal Production Department, Fac. of Agriculture, Assiut University, Assiut, Egypt
| |
Collapse
|
8
|
Enebe JT, Dim CC, Omeke AC. Maternal antioxidant micronutrient deficiencies among pre-eclamptic women in Enugu, Nigeria: a cross-sectional analytical study. J Int Med Res 2023; 51:3000605231209159. [PMID: 37940611 PMCID: PMC10637183 DOI: 10.1177/03000605231209159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/04/2023] [Indexed: 11/10/2023] Open
Abstract
OBJECTIVES To determine the prevalence of antioxidant micronutrient deficiencies in pregnant women with pre-eclampsia and healthy pregnant women, and to assess the relationships between trace element deficiency in pregnancy and the severity of pre-eclampsia in Enugu, Nigeria. METHODS We performed a secondary analysis of a cross-sectional analytical study of serum concentrations of copper, selenium, zinc, magnesium, and manganese in 81 pregnant women with pre-eclampsia and healthy pregnant women (controls) who were matched for age, gestational age, body mass index, and parity. This study was performed at the University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu. Participants' sera were analyzed with an atomic absorption spectrophotometer. RESULTS Trace element deficiencies were common and similar between women with pre-eclampsia and controls. However, women with pre-eclampsia were more likely to be deficient in manganese than controls (odds ratio = 2.28, 95% confidence interval: 1.90-2.75). Among the micronutrients studied, only manganese concentrations were significantly lower in women without severe symptoms of pre-eclampsia than in those with severe symptoms of pre-eclampsia. CONCLUSIONS Micronutrient deficiency is common in pregnant women with pre-eclampsia and in healthy pregnant women in Enugu, Nigeria. Only manganese deficiency is higher in women with pre-eclampsia than in healthy pregnant women.
Collapse
Affiliation(s)
- Joseph Tochukwu Enebe
- Department of Obstetrics and Gynaecology, Enugu State University of Science and Technology, College of Medicine/Teaching Hospital, Enugu, Nigeria
| | - Cyril Chukwudi Dim
- Department of Obstetrics and Gynaecology, College of Medicine, University of Nigeria Ituku/Ozalla Campus, Enugu state, Nigeria
| | - Akudo Chidimma Omeke
- Department of Obstetrics and Gynaecology, Enugu State University of Science and Technology, College of Medicine/Teaching Hospital, Enugu, Nigeria
| |
Collapse
|
9
|
Peila C, Riboldi L, Spada E, Coscia A, Barbagallo I, Li Volti G, Galvano F, Gazzolo D. The Gestational Pathologies Effect on the Human Milk Redox Homeostasis: A First Step towards Its Definition. Nutrients 2023; 15:4546. [PMID: 37960198 PMCID: PMC10648900 DOI: 10.3390/nu15214546] [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: 10/04/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Human Milk (HM) is a dynamic nourishment; its composition is influenced by several conditions such as gestational age, maternal diet and ethnicity. It appears important to evaluate the impact that gestational pathologies have on HM components and if their presence, as a source of oxidative stress in the mother, influence milk's redox homeostasis. To assess the effect of Preeclampsia (PE) and Gestational Diabetes Mellitus (GDM) on some aspects of human milk redox homeostasis, we chose to investigate both oxidative and antioxidant aspects, with, respectively, Lipid hydroperoxides (LOOHs) and Glutathione (GSH). METHODS Women with PE, GDM and who were healthy were recruited for this study. Colostrum, transitional and mature milk samples were collected. GSH and LOOHs levels were measured using a spectrophotometric test. To investigate the effect of pathology on redox homeostasis, a mixed linear model with unistructural covariance structure was performed. RESULTS A total of 120 mothers were recruited. The GSH concentration results were significantly lower in GDM women than in healthy women only in colostrum (p < 0.01). No other differences emerged. LOOHs was not detectable in almost all the samples. DISCUSSION Our study is the first to extensively evaluate these components in the HM of women with these gestational pathologies. The main observation is that GDM can alter the GSH level of HM, mainly in colostrum.
Collapse
Affiliation(s)
- Chiara Peila
- Neonatal Unit, Department of Public Health and Pediatrics, University of Turin, 10100 Turin, Italy
| | - Lorenzo Riboldi
- Neonatal Unit, Department of Public Health and Pediatrics, University of Turin, 10100 Turin, Italy
| | - Elena Spada
- Neonatal Unit, Department of Public Health and Pediatrics, University of Turin, 10100 Turin, Italy
| | - Alessandra Coscia
- Neonatal Unit, Department of Public Health and Pediatrics, University of Turin, 10100 Turin, Italy
| | - Ignazio Barbagallo
- Department of Biological Chemistry, Medical Chemistry and Molecular Biology, University of Catania, 95131 Catania, Italy
| | - Giovanni Li Volti
- Department of Biological Chemistry, Medical Chemistry and Molecular Biology, University of Catania, 95131 Catania, Italy
| | - Fabio Galvano
- Department of Biological Chemistry, Medical Chemistry and Molecular Biology, University of Catania, 95131 Catania, Italy
| | - Diego Gazzolo
- Neonatal Intensive Care Unit, University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
10
|
Coker SJ, Dyson RM, Smith-Díaz CC, Vissers MCM, Berry MJ. Effects of Low Vitamin C Intake on Fertility Parameters and Pregnancy Outcomes in Guinea Pigs. Nutrients 2023; 15:4107. [PMID: 37836389 PMCID: PMC10574174 DOI: 10.3390/nu15194107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Identifying how specific nutrients can impact fertility, pregnancy, and neonatal outcomes will yield important insights into the biological mechanisms linking diet and reproductive health. Our study investigates how dietary vitamin C intake affects various fertility parameters and pregnancy and neonatal outcomes in the guinea pig, a natural model of vitamin C dependency. Dunkin Hartley guinea pigs were fed an optimal (900 mg/kg feed) or low (100 mg/kg feed) vitamin C diet ad libitum for at least three weeks prior to mating and throughout pregnancy. We found that animals receiving the low vitamin C diet had an increased number of unsuccessful matings, a higher incidence of foetal reabsorption, and, among pregnancies resulting in delivery at term, produced fewer offspring. Neonates from mothers on the low vitamin C diet had significantly decreased plasma vitamin C concentrations at birth and exhibited mild growth impairments in a sex-dependent manner. We conclude that a diet low of vitamin C induces a state of subfertility, reduces overall fecundity, and adversely impacts both pregnancy outcomes and growth in the offspring. Our study provides an essential foundation for future investigations to determine whether these findings translate to humans. If so, they could have important clinical implications for assisted reproductive technologies and nutritional recommendations for couples trying to conceive, pregnant women, and breastfeeding mothers.
Collapse
Affiliation(s)
- Sharna J. Coker
- Perinatal and Developmental Physiology Group, Department of Paediatrics and Child Health, University of Otago, Wellington 6242, New Zealand; (S.J.C.); (R.M.D.)
| | - Rebecca M. Dyson
- Perinatal and Developmental Physiology Group, Department of Paediatrics and Child Health, University of Otago, Wellington 6242, New Zealand; (S.J.C.); (R.M.D.)
| | - Carlos C. Smith-Díaz
- Mātai Hāora—Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand;
| | - Margreet C. M. Vissers
- Mātai Hāora—Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand;
| | - Mary J. Berry
- Perinatal and Developmental Physiology Group, Department of Paediatrics and Child Health, University of Otago, Wellington 6242, New Zealand; (S.J.C.); (R.M.D.)
| |
Collapse
|
11
|
Mészáros B, Kukor Z, Valent S. Recent Advances in the Prevention and Screening of Preeclampsia. J Clin Med 2023; 12:6020. [PMID: 37762960 PMCID: PMC10532380 DOI: 10.3390/jcm12186020] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/08/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Throughout the history of medicine, preeclampsia has remained an enigmatic field of obstetrics. In 2023, despite its prevalence and impact, preeclampsia's exact cause and effective treatment remain elusive; the current options are limited to delivery. The purpose of this review is to summarize the knowledge of the possible novel prophylactic therapies and screening methods for preeclampsia, thereby providing valuable insights for healthcare professionals and researchers. Aspirin and LMWH have already been widely used; meanwhile, calcium, vitamin D, and pravastatin show promise, and endothelin receptor antagonists are being explored. Stress reduction, dietary changes, and lifestyle modifications are also being investigated. Another interesting and fast-growing area is AI- and software-based screening methods. It is also key to find novel biomarkers, which, in some cases, are not only able to predict the development of the disease, but some of them hold promise to be a potential therapeutic target. We conclude that, while a definitive cure for preeclampsia may not be eligible in the near future, it is likely that the assessment and enhancement of preventive methods will lead to the prevention of many cases. However, it is also important to highlight that more additional research is needed in the future to clarify the exact pathophysiology of preeclampsia and to thus identify potential therapeutic targets for more improved treatment methods.
Collapse
Affiliation(s)
- Balázs Mészáros
- Department of Obstetrics and Gynecology, Semmelweis University, 1082 Budapest, Hungary
| | - Zoltán Kukor
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, 1082 Budapest, Hungary
| | - Sándor Valent
- Department of Obstetrics and Gynecology, Semmelweis University, 1082 Budapest, Hungary
| |
Collapse
|
12
|
Alves PRMM, Fragoso MBT, Tenório MCS, Bueno NB, Goulart MOF, Oliveira ACM. The role played by oral antioxidant therapies in preventing and treating preeclampsia: An updated meta-analysis. Nutr Metab Cardiovasc Dis 2023; 33:1277-1292. [PMID: 37246073 DOI: 10.1016/j.numecd.2023.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 01/10/2023] [Accepted: 02/06/2023] [Indexed: 02/26/2023]
Abstract
AIMS Performing an up-to-date meta-analysis of oral antioxidant therapies and determining whether they are effective in preventing and/or treating preeclampsia (PE). DATA SYNTHESIS Search was performed in PubMed, CENTRAL, LILACS, Web of Science, and ScienceDirect databases. The risk of bias was assessed based on using Cochrane Collaboration's tool. A funnel plot was created, and Egger's and Peter's test was carried out to assess publication bias in the primary outcome of prevention studies. The overall quality of the evidence was assessed based on using the Grading of Recommendations Assessment, Developing and Evaluation (GRADE) tool; a formal protocol was published in the PROSPERO database (registration number CRD42022348992). In total, 32 studies were taken into consideration for analysis purposes; 22 studies focused on investigating preeclampsia prevention methods, whereas 10 focused on its treatment. Significant results associated with the incidence of preeclampsia were observed in prevention studies comprising 11,198 subjects and 1106 events in the control groups, as well as 11,156 subjects and 1048 events in the intervention groups (relative risk [RR]: 0.86, 95% confidence interval [CI]: [0.75, 0.99], P = 0.03; I2 = 44%, P = 0.02). With respect to outcomes associated with treatment studies, only intrauterine growth restriction has shown significant effects. Egger's and Peter's test has evidenced publication bias. Six outcomes in prevention studies were classified as having low quality and two as having moderate quality, whereas all three outcomes assessed in treatment studies were classified as having moderate quality. CONCLUSIONS Antioxidant therapy has shown beneficial effects on preeclampsia prevention; moreover, the positive impact of this therapy on intrauterine growth restriction was observed during the disease treatment.
Collapse
Affiliation(s)
- Palloma R M M Alves
- Faculdade de Nutrição, Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km 96,7, Tabuleiro dos Martins, CEP 57.072-970, Maceió, Alagoas, Brazil.
| | - Marilene B T Fragoso
- Instituto de Química e Biotecnologia (IQB/UFAL), Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km 96,7, Tabuleiro dos Martins, CEP 57.072-970, Maceió, Alagoas, Brazil; Rede Nordeste de Biotecnologia (RENORBIO), Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km 96,7, Tabuleiro dos Martins, CEP 57.072-970, Maceió, Alagoas, Brazil.
| | - Micaely C S Tenório
- Instituto de Química e Biotecnologia (IQB/UFAL), Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km 96,7, Tabuleiro dos Martins, CEP 57.072-970, Maceió, Alagoas, Brazil; Rede Nordeste de Biotecnologia (RENORBIO), Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km 96,7, Tabuleiro dos Martins, CEP 57.072-970, Maceió, Alagoas, Brazil.
| | - Nassib B Bueno
- Faculdade de Nutrição, Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km 96,7, Tabuleiro dos Martins, CEP 57.072-970, Maceió, Alagoas, Brazil.
| | - Marília O F Goulart
- Instituto de Química e Biotecnologia (IQB/UFAL), Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km 96,7, Tabuleiro dos Martins, CEP 57.072-970, Maceió, Alagoas, Brazil; Rede Nordeste de Biotecnologia (RENORBIO), Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km 96,7, Tabuleiro dos Martins, CEP 57.072-970, Maceió, Alagoas, Brazil.
| | - Alane C M Oliveira
- Faculdade de Nutrição, Universidade Federal de Alagoas, Campus A. C. Simões, BR 104 Norte, Km 96,7, Tabuleiro dos Martins, CEP 57.072-970, Maceió, Alagoas, Brazil.
| |
Collapse
|
13
|
Gunabalasingam S, De Almeida Lima Slizys D, Quotah O, Magee L, White SL, Rigutto-Farebrother J, Poston L, Dalrymple KV, Flynn AC. Micronutrient supplementation interventions in preconception and pregnant women at increased risk of developing pre-eclampsia: a systematic review and meta-analysis. Eur J Clin Nutr 2023; 77:710-730. [PMID: 36352102 PMCID: PMC10335932 DOI: 10.1038/s41430-022-01232-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Pre-eclampsia can lead to maternal and neonatal complications and is a common cause of maternal mortality worldwide. This review has examined the effect of micronutrient supplementation interventions in women identified as having a greater risk of developing pre-eclampsia. METHODS A systematic review was performed using the PRISMA guidelines. The electronic databases MEDLINE, EMBASE and the Cochrane Central Register of Controlled trials were searched for relevant literature and eligible studies identified according to a pre-specified criteria. A meta-analysis of randomised controlled trials (RCTs) was conducted to examine the effect of micronutrient supplementation on pre-eclampsia in high-risk women. RESULTS Twenty RCTs were identified and supplementation included vitamin C and E (n = 7), calcium (n = 5), vitamin D (n = 3), folic acid (n = 2), magnesium (n = 1) and multiple micronutrients (n = 2). Sample size and recruitment time point varied across studies and a variety of predictive factors were used to identify participants, with a previous history of pre-eclampsia being the most common. No studies utilised a validated prediction model. There was a reduction in pre-eclampsia with calcium (risk difference, -0.15 (-0.27, -0.03, I2 = 83.4%)), and vitamin D (risk difference, -0.09 (-0.17, -0.02, I2 = 0.0%)) supplementation. CONCLUSION Our findings show a lower rate of pre-eclampsia with calcium and vitamin D, however, conclusions were limited by small sample sizes, methodological variability and heterogeneity between studies. Further higher quality, large-scale RCTs of calcium and vitamin D are warranted. Exploration of interventions at different time points before and during pregnancy as well as those which utilise prediction modelling methodology, would provide greater insight into the efficacy of micronutrient supplementation intervention in the prevention of pre-eclampsia in high-risk women.
Collapse
Affiliation(s)
- Sowmiya Gunabalasingam
- Department of Women and Children's Health, School of Life Course and Population Sciences, King's College London, 10th Floor North Wing, St Thomas' Hospital, Westminster Bridge Road, London, SE1 7EH, UK
| | - Daniele De Almeida Lima Slizys
- Department of Women and Children's Health, School of Life Course and Population Sciences, King's College London, 10th Floor North Wing, St Thomas' Hospital, Westminster Bridge Road, London, SE1 7EH, UK
| | - Ola Quotah
- Department of Women and Children's Health, School of Life Course and Population Sciences, King's College London, 10th Floor North Wing, St Thomas' Hospital, Westminster Bridge Road, London, SE1 7EH, UK
| | - Laura Magee
- Department of Women and Children's Health, School of Life Course and Population Sciences, King's College London, 10th Floor North Wing, St Thomas' Hospital, Westminster Bridge Road, London, SE1 7EH, UK
| | - Sara L White
- Department of Women and Children's Health, School of Life Course and Population Sciences, King's College London, 10th Floor North Wing, St Thomas' Hospital, Westminster Bridge Road, London, SE1 7EH, UK
| | | | - Lucilla Poston
- Department of Women and Children's Health, School of Life Course and Population Sciences, King's College London, 10th Floor North Wing, St Thomas' Hospital, Westminster Bridge Road, London, SE1 7EH, UK
| | - Kathryn V Dalrymple
- Department of Population Health Sciences, School of Life Course and Population Sciences, King's College London, 4th floor Addison House, Guy's Campus, London, SE1 1UL, UK
| | - Angela C Flynn
- Department of Nutritional Sciences, School of Life Course and Population Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK.
| |
Collapse
|
14
|
Sheikh J, Allotey J, Kew T, Fernández-Félix BM, Zamora J, Khalil A, Thangaratinam S. Effects of race and ethnicity on perinatal outcomes in high-income and upper-middle-income countries: an individual participant data meta-analysis of 2 198 655 pregnancies. Lancet 2022; 400:2049-2062. [PMID: 36502843 DOI: 10.1016/s0140-6736(22)01191-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Existing evidence on the effects of race and ethnicity on pregnancy outcomes is restricted to individual studies done within specific countries and health systems. We aimed to assess the impact of race and ethnicity on perinatal outcomes in high-income and upper-middle-income countries, and to ascertain whether the magnitude of disparities, if any, varied across geographical regions. METHODS For this individual participant data (IPD) meta-analysis we used data from the International Prediction of Pregnancy Complications (IPPIC) Network of studies on pregnancy complications; the full dataset comprised 94 studies, 53 countries, and 4 539 640 pregnancies. We included studies that reported perinatal outcomes (neonatal death, stillbirth, preterm birth, and small-for-gestational-age babies) in at least two racial or ethnic groups (White, Black, south Asian, Hispanic, or other). For our two-step random-effects IPD meta-analysis, we did multiple imputations for confounder variables (maternal age, BMI, parity, and level of maternal education) selected with a directed acyclic graph. The primary outcomes were neonatal mortality and stillbirth. Secondary outcomes were preterm birth and a small-for-gestational-age baby. We estimated the association of race and ethnicity with perinatal outcomes using a multivariate logistic regression model and reported this association with odds ratios (ORs) and 95% CIs. We also did a subgroup analysis of studies by geographical region. FINDINGS 51 studies from 20 high-income and upper-middle-income countries, comprising 2 198 655 pregnancies, were eligible for inclusion in this IPD meta-analysis. Neonatal death was twice as likely in babies born to Black women than in babies born to White women (OR 2·00, 95% CI 1·44-2·78), as was stillbirth (2·16, 1·46-3·19), and babies born to Black women were at increased risk of preterm birth (1·65, 1·46-1·88) and being small for gestational age (1·39, 1·13-1·72). Babies of women categorised as Hispanic had a three-times increased risk of neonatal death (OR 3·34, 95% CI 2·77-4·02) than did those born to White women, and those born to south Asian women were at increased risk of preterm birth (OR 1·26, 95% CI 1·07-1·48) and being small for gestational age (1·61, 1·32-1·95). The effects of race and ethnicity on preterm birth and small-for-gestational-age babies did not vary across regions. INTERPRETATION Globally, among underserved groups, babies born to Black women had consistently poorer perinatal outcomes than White women after adjusting for maternal characteristics, although the risks varied for other groups. The effects of race and ethnicity on adverse perinatal outcomes did not vary by region. FUNDING National Institute for Health and Care Research, Wellbeing of Women.
Collapse
Affiliation(s)
- Jameela Sheikh
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - John Allotey
- WHO Collaborating Centre for Global Women's Health, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Tania Kew
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Borja M Fernández-Félix
- Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain; CIBER Epidemiology and Public Health, Madrid, Spain
| | - Javier Zamora
- WHO Collaborating Centre for Global Women's Health, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK; Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain; CIBER Epidemiology and Public Health, Madrid, Spain.
| | - Asma Khalil
- Foetal Medicine Unit, Department of Obstetrics and Gynaecology, St George's University Hospitals NHS Foundation Trust, London, UK; Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Shakila Thangaratinam
- WHO Collaborating Centre for Global Women's Health, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK; Birmingham Women's Hospital, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
15
|
Welch BM, McNell EE, Edin ML, Ferguson KK. Inflammation and oxidative stress as mediators of the impacts of environmental exposures on human pregnancy: Evidence from oxylipins. Pharmacol Ther 2022; 239:108181. [PMID: 35367517 PMCID: PMC9525454 DOI: 10.1016/j.pharmthera.2022.108181] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 02/08/2023]
Abstract
Inflammation and oxidative stress play major roles in healthy and pathological pregnancy. Environmental exposure to chemical pollutants may adversely affect maternal and fetal health in pregnancy by dysregulating these critical underlying processes of inflammation and oxidative stress. Oxylipins are bioactive lipids that play a major role in regulating inflammation and increasing lines of evidence point towards an importance in pregnancy. The biosynthetic production of oxylipins requires oxygenation of polyunsaturated fatty acids, which can occur through several well-characterized enzymatic and nonenzymatic pathways. This review describes the state of the science of epidemiologic evidence on oxylipin production in pregnancy and its association with 1) key pregnancy outcomes and 2) environmental exposures. We searched PubMed for studies of pregnancy that measured one or more oxylipin analytes during pregnancy or delivery. We evaluated oxylipin associations with three categories of adverse pregnancy outcomes, including preeclampsia, preterm birth, and fetal growth restriction, along with several categories of environmental pollutants. The majority of studies evaluated one to two oxylipins, most of which focused on oxylipins produced from nonenzymatic processes of oxidative stress. However, an increasing number of recent studies have leveraged technological advancements to profile a large number of oxylipins produced from distinct biosynthetic pathways. Although the literature indicated robust evidence that oxylipins produced via nonenzymatic pathways are associated with pregnancy outcomes and environmental exposures, evidence for enzymatically produced oxylipins showed that associations may differ between biosynthetic pathways. Along with summarizing this evidence, we review promising therapeutic options to regulate oxylipin production and provide a set of recommendations for future epidemiologic studies in these research areas. Further evidence is needed to improve our understanding of how oxylipins may act as key biological mediators for the adverse effects of environmental pollutants on pregnancy outcomes.
Collapse
Affiliation(s)
- Barrett M Welch
- Epidemiology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| | - Erin E McNell
- Epidemiology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Matthew L Edin
- Immunity, Inflammation, and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Kelly K Ferguson
- Epidemiology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| |
Collapse
|
16
|
Hamsir F, As’ad S, Tahir AM, Soraya D, Fujiko M, Chalid SMT, Idris I, Bukhari A, Hatta M, Bahar B, Efendi S. Macro- and Micronutrient of Junk Food and Preeclampsia on Pregnant Women. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.9949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Preeclampsia (PE) is still a global public health issue in developed and developing countries. It is a major contributor to maternal and fetal morbidity and mortality. The pathophysiology of preeclampsia during pregnancy is unclear, but the WHO reported that PE is higher in developing countries than developed ones due to lifestyle and nutritional status.
AIM: This study aimed to describe the influence of macro- and micronutrients of junk food on PE.
METHODS: This case–control study was carried out on 148 samples, including pregnant women with preeclampsia (n = 74) and normal pregnant women (n = 74) with gestational age > 20 weeks. Data on junk food intake were collected using the SQ-FFQ form and then estimated using the Nutri Survey. Data were analyzed in SPSS version 17 using descriptive statistics. Pearson correlation coefficient was used to observe the correlation between variables with p < 0.05 and 95% confidence interval.
RESULTS: Binary logistic regression revealed that the intake of fat (p = 0.005), vitamin C (p = 0.002), and sodium (p = 0.036) contained in the junk food were significantly associated with PE. The frequency of consumption of junk food also had a significant effect on the incidence of preeclampsia (p = 0.013).
CONCLUSION: Intake of fat, vitamin C, and sodium were associated with PE, and they were risk factors of PE. The frequency of consuming junk food gave more risk to have PE.
Collapse
|
17
|
Shi H, Jiang Y, Yuan P, Chen L, Gong X, Yang Y, Wang Y, Jiang H, Li Y, Sun M, Zhao Y, Wei Y. Association of Gestational Vitamin E Status With Pre-eclampsia: A Retrospective, Multicenter Cohort Study. Front Nutr 2022; 9:911337. [PMID: 35799589 PMCID: PMC9253635 DOI: 10.3389/fnut.2022.911337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
IntroductionPre-eclampsia is the second leading cause of maternal mortality worldwide. The controversy for the association of vitamin E with pre-eclampsia has raged unabated for two decades. We aimed to determine the association of vitamin E level in the first trimester and the gestational change with pre-eclampsia.Materials and MethodsA retrospective cohort study was conducted among singleton pregnant women aged 15–49 years at 137 hospitals in China. Serum vitamin E concentrations in the first trimester and at pre-eclampsia assessment time were uniformly quantified in a laboratory by high performance liquid chromatography. Logistic regression models with restricted cubic splines were performed to reveal a non-linear association of vitamin E concentrations in the first trimester and the gestational change with pre-eclampsia.ResultsWe included 73 317 participants (47.8% aged 25–29 years) and 2.28% were diagnosed with pre-eclampsia. Higher risk was observed in those with lower concentration in the first trimester and greater gestational decrease, with a range from 0.81 to 80.60%. A non-linear L-shaped association was observed between vitamin E concentrations in the first trimester and pre-eclampsia, suggesting a threshold at 7.3 mg/L and a ceiling effect: the risk saw a steep rise when the concentrations in the first trimester were < 7.3 mg/L but was relatively flat beyond the inflection point. Sharply increased pre-eclampsia risk was also found in those with gestational vitamin E decrease after accounting for the baseline status in the first trimester. However, gestational vitamin E increase was associated with decreased pre-eclampsia risk when the baseline concentrations were < 7.3 mg/L but did not confer additional benefits when it was above the threshold.ConclusionWe demonstrated alarmingly high pre-eclampsia risk in women with vitamin E concentrations of < 7.3 mg/L in the first trimester and gestational vitamin E decrease. These findings underscore the need to supplement vitamin E among pregnant women with low baseline status.
Collapse
Affiliation(s)
- Huifeng Shi
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Centre for Obstetrics and Gynecology, Beijing, China
- National Centre for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Yuanhui Jiang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Centre for Obstetrics and Gynecology, Beijing, China
| | - Pengbo Yuan
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Centre for Obstetrics and Gynecology, Beijing, China
| | - Lian Chen
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Centre for Obstetrics and Gynecology, Beijing, China
- National Centre for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Xiaoli Gong
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Centre for Obstetrics and Gynecology, Beijing, China
| | - Yike Yang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Centre for Obstetrics and Gynecology, Beijing, China
| | - Yuanyuan Wang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Centre for Obstetrics and Gynecology, Beijing, China
| | - Hai Jiang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Centre for Obstetrics and Gynecology, Beijing, China
| | - You Li
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Centre for Obstetrics and Gynecology, Beijing, China
| | - Mengxing Sun
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Centre for Obstetrics and Gynecology, Beijing, China
| | - Yangyu Zhao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Centre for Obstetrics and Gynecology, Beijing, China
- National Centre for Healthcare Quality Management in Obstetrics, Beijing, China
- Yangyu Zhao,
| | - Yuan Wei
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Centre for Obstetrics and Gynecology, Beijing, China
- National Centre for Healthcare Quality Management in Obstetrics, Beijing, China
- *Correspondence: Yuan Wei,
| |
Collapse
|
18
|
Bigham Z, Robles Y, Freund KM, Palmer JR, Bertrand KA. Hypertensive diseases of pregnancy and risk of breast cancer in the Black Women's Health Study. Breast Cancer Res Treat 2022; 194:127-135. [PMID: 35478297 DOI: 10.1007/s10549-022-06606-3] [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] [Received: 12/15/2021] [Accepted: 04/06/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE Compared to white women, Black women have increased risk of developing hypertensive diseases of pregnancy (HDOP) and have a higher incidence of aggressive breast cancer subtypes. Few studies of HDOP and breast cancer risk have included large numbers of Black women. This study examined the relation of HDOP to incidence of breast cancer overall and by estrogen receptor (ER) status in Black women. METHODS We followed 42,982 parous women in the Black Women's Health Study, a nationwide prospective study of Black women. Multivariable Cox proportional hazards regression models were used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) to assess associations of self-reported HDOP, including preeclampsia and gestational hypertension, with breast cancer incidence overall and by ER subtype, adjusted for age and established breast cancer risk factors. RESULTS Over 20 years of follow-up, we identified 2376 incident breast cancer cases. History of HDOP (11.7%) was not associated with breast cancer risk overall (HR 0.98; 95% CI 0.87, 1.11). HRs for invasive ER+ and ER- breast cancer were 1.11 (95% CI 0.93, 1.34) and 0.81 (95% CI 0.61, 1.07), respectively. CONCLUSIONS HDOP was not associated with risk of overall breast cancer in Black women. A suggestive inverse association with ER- breast cancer may reflect an anti-tumorigenic hormone profile in HDOP, but those results require confirmation in other studies.
Collapse
Affiliation(s)
- Zahna Bigham
- Tufts University School of Medicine and Graduate School of Biomedical Sciences, Boston, MA, USA
| | - Yvonne Robles
- Slone Epidemiology Center at Boston University, 72 East Concord Street, L-7, Boston, MA, USA
| | - Karen M Freund
- Tufts University School of Medicine and Graduate School of Biomedical Sciences, Boston, MA, USA.,Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Julie R Palmer
- Slone Epidemiology Center at Boston University, 72 East Concord Street, L-7, Boston, MA, USA
| | - Kimberly A Bertrand
- Slone Epidemiology Center at Boston University, 72 East Concord Street, L-7, Boston, MA, USA.
| |
Collapse
|
19
|
Di Fabrizio C, Giorgione V, Khalil A, Murdoch CE. Antioxidants in Pregnancy: Do We Really Need More Trials? Antioxidants (Basel) 2022; 11:812. [PMID: 35624676 PMCID: PMC9137466 DOI: 10.3390/antiox11050812] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 02/01/2023] Open
Abstract
Human pregnancy can be affected by numerous pathologies, from those which are mild and reversible to others which are life-threatening. Among these, gestational diabetes mellitus and hypertensive disorders of pregnancy with subsequent consequences stand out. Health problems experienced by women during pregnancy and postpartum are associated with significant costs to health systems worldwide and contribute largely to maternal mortality and morbidity. Major risk factors for mothers include obesity, advanced maternal age, cardiovascular dysfunction, and endothelial damage; in these scenarios, oxidative stress plays a major role. Markers of oxidative stress can be measured in patients with preeclampsia, foetal growth restriction, and gestational diabetes mellitus, even before their clinical onset. In consequence, antioxidant supplements have been proposed as a possible therapy; however, results derived from large scale randomised clinical trials have been disappointing as no positive effects were demonstrated. This review focuses on the latest evidence on oxidative stress in pregnancy complications, their early diagnosis, and possible therapies to prevent or treat these pathologies.
Collapse
Affiliation(s)
- Carolina Di Fabrizio
- Vascular Biology Research Center, Molecular and Clinical Sciences Research Institute, St George’s University of London, London SW17 0QT, UK; (C.D.F.); (V.G.); (A.K.)
- Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Veronica Giorgione
- Vascular Biology Research Center, Molecular and Clinical Sciences Research Institute, St George’s University of London, London SW17 0QT, UK; (C.D.F.); (V.G.); (A.K.)
| | - Asma Khalil
- Vascular Biology Research Center, Molecular and Clinical Sciences Research Institute, St George’s University of London, London SW17 0QT, UK; (C.D.F.); (V.G.); (A.K.)
- Fetal Medicine Unit, St George’s University Hospitals NHS Foundation Trust, London SW17 0QT, UK
| | - Colin E. Murdoch
- Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| |
Collapse
|
20
|
Manna S, Ruano CSM, Hegenbarth JC, Vaiman D, Gupta S, McCarthy FP, Méhats C, McCarthy C, Apicella C, Scheel J. Computational Models on Pathological Redox Signalling Driven by Pregnancy: A Review. Antioxidants (Basel) 2022; 11:585. [PMID: 35326235 PMCID: PMC8945226 DOI: 10.3390/antiox11030585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 02/04/2023] Open
Abstract
Oxidative stress is associated with a myriad of diseases including pregnancy pathologies with long-term cardiovascular repercussions for both the mother and baby. Aberrant redox signalling coupled with deficient antioxidant defence leads to chronic molecular impairment. Abnormal placentation has been considered the primary source for reactive species; however, placental dysfunction has been deemed secondary to maternal cardiovascular maladaptation in pregnancy. While various therapeutic interventions, aimed at combating deregulated oxidative stress during pregnancy have shown promise in experimental models, they often result as inconclusive or detrimental in clinical trials, warranting the need for further research to identify candidates. The strengths and limitations of current experimental methods in redox research are discussed. Assessment of redox status and oxidative stress in experimental models and in clinical practice remains challenging; the state-of-the-art of computational models in this field is presented in this review, comparing static and dynamic models which provide functional information such as protein-protein interactions, as well as the impact of changes in molecular species on the redox-status of the system, respectively. Enhanced knowledge of redox biology in during pregnancy through computational modelling such as generation of Systems Biology Markup Language model which integrates existing models to a larger network in the context of placenta physiology.
Collapse
Affiliation(s)
- Samprikta Manna
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, University College Cork, T12 YE02 Cork, Ireland;
| | - Camino S. M. Ruano
- Institut Cochin, Inserm U1016, UMR8104 CNRS, Université de Paris, 75014 Paris, France; (C.S.M.R.); (D.V.); (C.M.); (C.A.)
| | - Jana-Charlotte Hegenbarth
- Department of Molecular Genetics, Faculty of Science and Engineering, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 KH Maastricht, The Netherlands;
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Daniel Vaiman
- Institut Cochin, Inserm U1016, UMR8104 CNRS, Université de Paris, 75014 Paris, France; (C.S.M.R.); (D.V.); (C.M.); (C.A.)
| | - Shailendra Gupta
- Department of Systems Biology and Bioinformatics, Rostock University, 18051 Rostock, Germany; (S.G.); (J.S.)
| | - Fergus P. McCarthy
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, University College Cork, T12 YE02 Cork, Ireland;
| | - Céline Méhats
- Institut Cochin, Inserm U1016, UMR8104 CNRS, Université de Paris, 75014 Paris, France; (C.S.M.R.); (D.V.); (C.M.); (C.A.)
| | - Cathal McCarthy
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, T12 K8AF Cork, Ireland;
| | - Clara Apicella
- Institut Cochin, Inserm U1016, UMR8104 CNRS, Université de Paris, 75014 Paris, France; (C.S.M.R.); (D.V.); (C.M.); (C.A.)
| | - Julia Scheel
- Department of Systems Biology and Bioinformatics, Rostock University, 18051 Rostock, Germany; (S.G.); (J.S.)
| |
Collapse
|
21
|
Md Amin NA, Sheikh Abdul Kadir SH, Arshad AH, Abdul Aziz N, Abdul Nasir NA, Ab Latip N. Are Vitamin E Supplementation Beneficial for Female Gynaecology Health and Diseases? Molecules 2022; 27:molecules27061896. [PMID: 35335260 PMCID: PMC8955126 DOI: 10.3390/molecules27061896] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/06/2022] [Accepted: 03/10/2022] [Indexed: 12/04/2022] Open
Abstract
Vitamin E is known as an essential vitamin, and many studies had demonstrated the importance of vitamin E throughout the reproductive process, such as miscarriage, premature birth, preeclampsia, and intrauterine growth restriction, which could be caused by a lack of vitamin E during pregnancy. Its potent antioxidant properties can counteract the oxidative stress induced by oxygen free radicals and imbalance of oxidative-antioxidant levels, hence it may play a role in maintaining the normal function of the female reproductive system. Despite the fact that vitamin E is acknowledged as the substance needed for reproduction, its beneficial effects on female fertility, gynaecological health, and diseases are still poorly understood and lacking. Therefore, the goal of this paper is to provide a summary of the known roles of vitamin E supplementation in women for gynaecological health and reproductive-related diseases, as well as its future perspective.
Collapse
Affiliation(s)
- Nur Amira Md Amin
- Institute for Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA, Cawangan Selangor, Sungai Buloh 47000, Selangor, Malaysia;
- Institute of Medical Molecular Biotechnology (IMMB), Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Sungai Buloh 47000, Selangor, Malaysia
- Department of Biochemistry, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Sungai Buloh 47000, Selangor, Malaysia
| | - Siti Hamimah Sheikh Abdul Kadir
- Institute for Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA, Cawangan Selangor, Sungai Buloh 47000, Selangor, Malaysia;
- Institute of Medical Molecular Biotechnology (IMMB), Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Sungai Buloh 47000, Selangor, Malaysia
- Department of Biochemistry, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Sungai Buloh 47000, Selangor, Malaysia
- Correspondence:
| | - Akmal Hisyam Arshad
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh 47000, Selangor, Malaysia;
| | - Norhaslinda Abdul Aziz
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras 56000, Kuala Lumpur, Malaysia;
| | - Nurul Alimah Abdul Nasir
- Department of Pharmacology, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Sungai Buloh 47000, Selangor, Malaysia;
| | - Normala Ab Latip
- Atta-ur-Rahman Institute for Natural Products Discovery (AuRIns), Faculty of Pharmacy, Universiti Teknologi MARA, Cawangan Selangor, Puncak Alam 42300, Selangor, Malaysia;
| |
Collapse
|
22
|
Tong S, Kaitu’u-Lino TJ, Hastie R, Brownfoot F, Cluver C, Hannan N. Pravastatin, proton-pump inhibitors, metformin, micronutrients, and biologics: new horizons for the prevention or treatment of preeclampsia. Am J Obstet Gynecol 2022; 226:S1157-S1170. [PMID: 32946849 DOI: 10.1016/j.ajog.2020.09.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/25/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022]
Abstract
There has been increasing research momentum to identify new therapeutic agents for the prevention or treatment of preeclampsia, drugs that can affect the underlying disease pathophysiology. Molecular targets of candidate treatments include oxidative stress, antiangiogenic factors, and the angiotensin, nitric oxide, and proinflammatory pathways. The proposed treatments undergoing preclinical and clinical trial evaluation are thought to act on placental or endothelial disease or both. Most have adopted the pragmatic strategy of repurposing drugs. Of all the therapeutic agents proposed, pravastatin has received the most interest. There are preclinical studies showing that it has pleiotropic actions that favorably impact on multiple molecular targets and can resolve a preeclampsia phenotype in many animal models. An early phase clinical trial suggests that it may have therapeutic activity. Several large prevention trials are planned or ongoing and, when completed, could definitively address whether pravastatin can prevent preeclampsia. Proton-pump inhibitors, metformin, and sulfasalazine are other drugs with preclinical evidence of multiple molecular actions that could resolve the pathophysiology of preeclampsia. These agents are also currently being evaluated in clinical trials. There have been many recent preclinical studies identifying the potential of numerous natural compounds to treat preeclampsia, such as plant extracts and micronutrients that have potent anti-inflammatory or antioxidant activity. Recent preclinical studies have also proposed novel molecular-targeted strategies, such as monoclonal antibodies targeting tumor necrosis factor alpha, placental growth factor, and short interfering RNA technology, to silence the gene expression of soluble fms-like tyrosine kinase-1 or angiotensinogen. Other treatment approaches that have transitioned to human trials (ranging from single-arm to phase III trials that have been completed or are ongoing) include folic acid, nitric oxide donors (such as L-arginine), recombinant antithrombin III, digoxin immune antigen-binding fragment, and melatonin. There have been case series showing the removal of circulating soluble fms-like tyrosine kinase-1 may help stabilize the disease and prolong pregnancy. Interestingly, there are case reports suggesting that monoclonal antibody eculizumab (complement inhibitor) may have therapeutic potential. If new agents are discovered that are proven to be effective in preventing or treating preeclampsia, the potential to improve global maternal and perinatal health will be significant.
Collapse
|
23
|
Redman CW, Staff AC, Roberts JM. Syncytiotrophoblast stress in preeclampsia: the convergence point for multiple pathways. Am J Obstet Gynecol 2022; 226:S907-S927. [PMID: 33546842 DOI: 10.1016/j.ajog.2020.09.047] [Citation(s) in RCA: 183] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/14/2020] [Accepted: 09/19/2020] [Indexed: 12/29/2022]
Abstract
Preeclampsia evolves in 2 stages: a placental problem that generates signals to the mother to cause a range of responses that comprise the second stage (preeclampsia syndrome). The first stage of early-onset preeclampsia is poor placentation, which we here call malplacentation. The spiral arteries are incompletely remodeled, leading to later placental malperfusion, relatively early in the second half of pregnancy. The long duration of the first stage (several months) is unsurprisingly associated with fetal growth restriction. The first stage of late-onset preeclampsia, approximately 80% of total cases, is shorter (several weeks) and part of a process that is common to all pregnancies. Placental function declines as it outgrows uterine capacity, with increasing chorionic villous packing, compression of the intervillous space, and fetal hypoxia, and causes late-onset clinical presentations such as "unexplained" stillbirths, late-onset fetal growth restriction, or preeclampsia. The second stages of early- and late-onset preeclampsia share syncytiotrophoblast stress as the most relevant feature that causes the maternal syndrome. Syncytiotrophoblast stress signals in the maternal circulation are probably the most specific biomarkers for preeclampsia. In addition, soluble fms-like tyrosine kinase-1 (mainly produced by syncytiotrophoblast) is the best-known biomarker and is routinely used in clinical practice in many locations. How the stress signals change over time in normal pregnancies indicates that syncytiotrophoblast stress begins on average at 30 to 32 weeks' gestation and progresses to term. At term, syncytiotrophoblast shows increasing markers of stress, including apoptosis, pyroptosis, autophagy, syncytial knots, and necrosis. We label this phenotype the "twilight placenta" and argue that it accounts for the clinical problems of postmature pregnancies. Senescence as a stress response differs in multinuclear syncytiotrophoblast from that of mononuclear cells. Syncytiotrophoblast irreversibly acquires part of the senescence phenotype (cell cycle arrest) when it is formed by cell fusion. The 2 pathways converge on the common pathologic endpoint, syncytiotrophoblast stress, and contribute to preeclampsia subtypes. We highlight that the well-known heterogeneity of the preeclampsia syndrome arises from different pathways to this common endpoint, influenced by maternal genetics, epigenetics, lifestyle, and environmental factors with different fetal and maternal responses to the ensuing insults. This complexity mandates a reassessment of our approach to predicting and preventing preeclampsia, and we summarize research priorities to maximize what we can learn about these important issues.
Collapse
|
24
|
Aye IL, Aiken CE, Charnock-Jones DS, Smith GC. Placental energy metabolism in health and disease-significance of development and implications for preeclampsia. Am J Obstet Gynecol 2022; 226:S928-S944. [PMID: 33189710 DOI: 10.1016/j.ajog.2020.11.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 02/08/2023]
Abstract
The placenta is a highly metabolically active organ fulfilling the bioenergetic and biosynthetic needs to support its own rapid growth and that of the fetus. Placental metabolic dysfunction is a common occurrence in preeclampsia although its causal relationship to the pathophysiology is unclear. At the outset, this may simply be seen as an "engine out of fuel." However, placental metabolism plays a vital role beyond energy production and is linked to physiological and developmental processes. In this review, we discuss the metabolic basis for placental dysfunction and propose that the alterations in energy metabolism may explain many of the placental phenotypes of preeclampsia such as reduced placental and fetal growth, redox imbalance, oxidative stress, altered epigenetic and gene expression profiles, and the functional consequences of these aberrations. We propose that placental metabolic reprogramming reflects the dynamic physiological state allowing the tissue to adapt to developmental changes and respond to preeclampsia stress, whereas the inability to reprogram placental metabolism may result in severe preeclampsia phenotypes. Finally, we discuss common tested and novel therapeutic strategies for treating placental dysfunction in preeclampsia and their impact on placental energy metabolism as possible explanations into their potential benefits or harm.
Collapse
|
25
|
Coker SJ, Smith-Díaz CC, Dyson RM, Vissers MCM, Berry MJ. The Epigenetic Role of Vitamin C in Neurodevelopment. Int J Mol Sci 2022; 23:ijms23031208. [PMID: 35163133 PMCID: PMC8836017 DOI: 10.3390/ijms23031208] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 02/07/2023] Open
Abstract
The maternal diet during pregnancy is a key determinant of offspring health. Early studies have linked poor maternal nutrition during gestation with a propensity for the development of chronic conditions in offspring. These conditions include cardiovascular disease, type 2 diabetes and even compromised mental health. While multiple factors may contribute to these outcomes, disturbed epigenetic programming during early development is one potential biological mechanism. The epigenome is programmed primarily in utero, and during this time, the developing fetus is highly susceptible to environmental factors such as nutritional insults. During neurodevelopment, epigenetic programming coordinates the formation of primitive central nervous system structures, neurogenesis, and neuroplasticity. Dysregulated epigenetic programming has been implicated in the aetiology of several neurodevelopmental disorders such as Tatton-Brown-Rahman syndrome. Accordingly, there is great interest in determining how maternal nutrient availability in pregnancy might affect the epigenetic status of offspring, and how such influences may present phenotypically. In recent years, a number of epigenetic enzymes that are active during embryonic development have been found to require vitamin C as a cofactor. These enzymes include the ten-eleven translocation methylcytosine dioxygenases (TETs) and the Jumonji C domain-containing histone lysine demethylases that catalyse the oxidative removal of methyl groups on cytosines and histone lysine residues, respectively. These enzymes are integral to epigenetic regulation and have fundamental roles in cellular differentiation, the maintenance of pluripotency and development. The dependence of these enzymes on vitamin C for optimal catalytic activity illustrates a potentially critical contribution of the nutrient during mammalian development. These insights also highlight a potential risk associated with vitamin C insufficiency during pregnancy. The link between vitamin C insufficiency and development is particularly apparent in the context of neurodevelopment and high vitamin C concentrations in the brain are indicative of important functional requirements in this organ. Accordingly, this review considers the evidence for the potential impact of maternal vitamin C status on neurodevelopmental epigenetics.
Collapse
Affiliation(s)
- Sharna J. Coker
- Perinatal & Developmental Physiology Group, Department of Paediatrics & Child Health, University of Otago, Wellington 6242, New Zealand; (S.J.C.); (R.M.D.)
| | - Carlos C. Smith-Díaz
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand;
| | - Rebecca M. Dyson
- Perinatal & Developmental Physiology Group, Department of Paediatrics & Child Health, University of Otago, Wellington 6242, New Zealand; (S.J.C.); (R.M.D.)
| | - Margreet C. M. Vissers
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand;
- Correspondence: (M.C.M.V.); (M.J.B.)
| | - Mary J. Berry
- Perinatal & Developmental Physiology Group, Department of Paediatrics & Child Health, University of Otago, Wellington 6242, New Zealand; (S.J.C.); (R.M.D.)
- Correspondence: (M.C.M.V.); (M.J.B.)
| |
Collapse
|
26
|
Head B, Traber MG. Expanding role of vitamin E in protection against metabolic dysregulation: Insights gained from model systems, especially the developing nervous system of zebrafish embryos. Free Radic Biol Med 2021; 176:80-91. [PMID: 34555455 DOI: 10.1016/j.freeradbiomed.2021.09.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/27/2021] [Accepted: 09/07/2021] [Indexed: 12/20/2022]
Abstract
This review discusses why the embryo requires vitamin E (VitE) and shows that its lack causes metabolic dysregulation and impacts morphological changes at very early stages in development, which occur prior to when a woman knows she is pregnant. VitE halts the chain reactions of lipid peroxidation (LPO). Metabolomic analyses indicate that thiols become depleted in E- embryos because LPO generates products that require compensation using limited amino acids and methyl donors that are also developmentally relevant. Thus, VitE protects metabolic networks and the integrated gene expression networks that control development. VitE is critical especially for neurodevelopment, which is dependent on trafficking by the α-tocopherol transfer protein (TTPa). VitE-deficient (E-) zebrafish embryos initially appear normal, but by 12 and 24 h post-fertilization (hpf) E- embryos are developmentally abnormal with expression of pax2a and sox10 mis-localized in the midbrain-hindbrain boundary, neural crest cells and throughout the spinal neurons. These patterning defects indicate cells that are especially in need of VitE-protection. They precede obvious morphological abnormalities (cranial-facial malformation, pericardial edema, yolksac edema, skewed body-axis) and impaired behavioral responses to locomotor activity tests. The TTPA gene (ttpa) is expressed at the leading edges of the brain ventricle border. Ttpa knockdown using morpholinos is 100% lethal by 24 hpf, while E- embryo brains are often over- or under-inflated at 24 hpf. Further, E- embryos prior to 24 hpf have increased expression of genes involved in glycolysis and the pentose phosphate pathway, and decreased expression of genes involved in anabolic pathways and transcription. Combined data from both gene expression and the metabolome in E- embryos at 24 hpf suggest that the activity of the mechanistic Target of Rapamycin (mTOR) signaling pathway is decreased, which may impact both metabolism and neurodevelopment. Further evaluation of VitE deficiency in neurogenesis and its subsequent impact on learning and behavior is needed.
Collapse
Affiliation(s)
- Brian Head
- Linus Pauling Institute, Corvallis, OR, USA; Molecular and Cell Biology Program, Corvallis, OR, USA
| | - Maret G Traber
- Linus Pauling Institute, Corvallis, OR, USA; School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA.
| |
Collapse
|
27
|
Allotey J, Snell KI, Smuk M, Hooper R, Chan CL, Ahmed A, Chappell LC, von Dadelszen P, Dodds J, Green M, Kenny L, Khalil A, Khan KS, Mol BW, Myers J, Poston L, Thilaganathan B, Staff AC, Smith GC, Ganzevoort W, Laivuori H, Odibo AO, Ramírez JA, Kingdom J, Daskalakis G, Farrar D, Baschat AA, Seed PT, Prefumo F, da Silva Costa F, Groen H, Audibert F, Masse J, Skråstad RB, Salvesen KÅ, Haavaldsen C, Nagata C, Rumbold AR, Heinonen S, Askie LM, Smits LJ, Vinter CA, Magnus PM, Eero K, Villa PM, Jenum AK, Andersen LB, Norman JE, Ohkuchi A, Eskild A, Bhattacharya S, McAuliffe FM, Galindo A, Herraiz I, Carbillon L, Klipstein-Grobusch K, Yeo S, Teede HJ, Browne JL, Moons KG, Riley RD, Thangaratinam S. Validation and development of models using clinical, biochemical and ultrasound markers for predicting pre-eclampsia: an individual participant data meta-analysis. Health Technol Assess 2021; 24:1-252. [PMID: 33336645 DOI: 10.3310/hta24720] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Pre-eclampsia is a leading cause of maternal and perinatal mortality and morbidity. Early identification of women at risk is needed to plan management. OBJECTIVES To assess the performance of existing pre-eclampsia prediction models and to develop and validate models for pre-eclampsia using individual participant data meta-analysis. We also estimated the prognostic value of individual markers. DESIGN This was an individual participant data meta-analysis of cohort studies. SETTING Source data from secondary and tertiary care. PREDICTORS We identified predictors from systematic reviews, and prioritised for importance in an international survey. PRIMARY OUTCOMES Early-onset (delivery at < 34 weeks' gestation), late-onset (delivery at ≥ 34 weeks' gestation) and any-onset pre-eclampsia. ANALYSIS We externally validated existing prediction models in UK cohorts and reported their performance in terms of discrimination and calibration. We developed and validated 12 new models based on clinical characteristics, clinical characteristics and biochemical markers, and clinical characteristics and ultrasound markers in the first and second trimesters. We summarised the data set-specific performance of each model using a random-effects meta-analysis. Discrimination was considered promising for C-statistics of ≥ 0.7, and calibration was considered good if the slope was near 1 and calibration-in-the-large was near 0. Heterogeneity was quantified using I 2 and τ2. A decision curve analysis was undertaken to determine the clinical utility (net benefit) of the models. We reported the unadjusted prognostic value of individual predictors for pre-eclampsia as odds ratios with 95% confidence and prediction intervals. RESULTS The International Prediction of Pregnancy Complications network comprised 78 studies (3,570,993 singleton pregnancies) identified from systematic reviews of tests to predict pre-eclampsia. Twenty-four of the 131 published prediction models could be validated in 11 UK cohorts. Summary C-statistics were between 0.6 and 0.7 for most models, and calibration was generally poor owing to large between-study heterogeneity, suggesting model overfitting. The clinical utility of the models varied between showing net harm to showing minimal or no net benefit. The average discrimination for IPPIC models ranged between 0.68 and 0.83. This was highest for the second-trimester clinical characteristics and biochemical markers model to predict early-onset pre-eclampsia, and lowest for the first-trimester clinical characteristics models to predict any pre-eclampsia. Calibration performance was heterogeneous across studies. Net benefit was observed for International Prediction of Pregnancy Complications first and second-trimester clinical characteristics and clinical characteristics and biochemical markers models predicting any pre-eclampsia, when validated in singleton nulliparous women managed in the UK NHS. History of hypertension, parity, smoking, mode of conception, placental growth factor and uterine artery pulsatility index had the strongest unadjusted associations with pre-eclampsia. LIMITATIONS Variations in study population characteristics, type of predictors reported, too few events in some validation cohorts and the type of measurements contributed to heterogeneity in performance of the International Prediction of Pregnancy Complications models. Some published models were not validated because model predictors were unavailable in the individual participant data. CONCLUSION For models that could be validated, predictive performance was generally poor across data sets. Although the International Prediction of Pregnancy Complications models show good predictive performance on average, and in the singleton nulliparous population, heterogeneity in calibration performance is likely across settings. FUTURE WORK Recalibration of model parameters within populations may improve calibration performance. Additional strong predictors need to be identified to improve model performance and consistency. Validation, including examination of calibration heterogeneity, is required for the models we could not validate. STUDY REGISTRATION This study is registered as PROSPERO CRD42015029349. FUNDING This project was funded by the National Institute for Health Research (NIHR) Health Technology Assessment programme and will be published in full in Health Technology Assessment; Vol. 24, No. 72. See the NIHR Journals Library website for further project information.
Collapse
|
28
|
Lofthouse EM, Manousopoulou A, Cleal JK, O'Kelly IM, Poore KR, Garbis SD, Lewis RM. N-acetylcysteine, xCT and suppression of Maxi-chloride channel activity in human placenta. Placenta 2021; 110:46-55. [PMID: 34120018 DOI: 10.1016/j.placenta.2021.05.009] [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: 03/05/2021] [Revised: 05/04/2021] [Accepted: 05/26/2021] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Placental oxidative stress features in pregnancy pathologies but in clinical trials antioxidant supplementation has not improved outcomes. N-acetylcysteine (NAC) stimulates glutathione production and is proposed as a therapeutic agent in pregnancy. However, key elements of N-acetylcysteine biology, including its cellular uptake mechanism, remains unclear. This study explores how the cystine/glutamate transporter xCT may mediate N-acetylcysteine uptake and how N-acetylcysteine alters placental redox status. METHODS The involvement of xCT in NAC uptake by the human placenta was studied in perfused placenta and Xenopus oocytes. The effect of short-term N-acetylcysteine exposure on the placental villous proteome was determined using LC-MS. The effect of N-acetylcysteine on Maxi-chloride channel activity was investigated in perfused placenta, villous fragments and cell culture. RESULTS Maternoplacental N-acetylcysteine administration stimulated intracellular glutamate efflux suggesting a role of the exchange transporter xCT, which was localised to the microvillous membrane of the placental syncytiotrophoblast. Placental exposure to a bolus of N-acetylcysteine inhibited subsequent activation of the redox sensitive Maxi-chloride channel independently of glutathione synthesis. Stable isotope quantitative proteomics of placental villi treated with N-acetylcysteine demonstrated changes in pathways associated with oxidative stress, apoptosis and the acute phase response. DISCUSSION This study suggests that xCT mediates N-acetylcysteine uptake into the placenta and that N-acetylcysteine treatment of placental tissue alters the placental proteome while regulating the redox sensitive Maxi-chloride channel. Interestingly N-acetylcysteine had antioxidant effects independent of the glutathione pathway. Effective placental antioxidant therapy in pregnancy may require maintaining the balance between normalising redox status without inhibiting physiological redox signalling.
Collapse
Affiliation(s)
- Emma M Lofthouse
- Faculty of Medicine, University of Southampton, UK; Institute for Life Sciences, University of Southampton, UK.
| | - Antigoni Manousopoulou
- Faculty of Medicine, University of Southampton, UK; Institute for Life Sciences, University of Southampton, UK; Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Jane K Cleal
- Faculty of Medicine, University of Southampton, UK; Institute for Life Sciences, University of Southampton, UK
| | | | | | - Spiros D Garbis
- Faculty of Medicine, University of Southampton, UK; Institute for Life Sciences, University of Southampton, UK; Proteome Exploration Laboratory, Beckman Institute, Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Rohan M Lewis
- Faculty of Medicine, University of Southampton, UK; Institute for Life Sciences, University of Southampton, UK
| |
Collapse
|
29
|
Keats EC, Oh C, Chau T, Khalifa DS, Imdad A, Bhutta ZA. Effects of vitamin and mineral supplementation during pregnancy on maternal, birth, child health and development outcomes in low- and middle-income countries: A systematic review. CAMPBELL SYSTEMATIC REVIEWS 2021; 17:e1127. [PMID: 37051178 PMCID: PMC8356361 DOI: 10.1002/cl2.1127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Background Almost two billion people who are deficient in vitamins and minerals are women and children in low- and middle-income countries (LMIC). These deficiencies are worsened during pregnancy due to increased energy and nutritional demands, causing adverse outcomes in mother and child. To reduce micronutrient deficiencies, several strategies have been implemented, including diet diversification, large-scale and targeted fortification, staple crop bio-fortification and micronutrient supplementation. Objectives To evaluate and summarize the available evidence on the effects of micronutrient supplementation during pregnancy in LMIC on maternal, fetal, child health and child development outcomes. This review will assess the impact of single micronutrient supplementation (calcium, vitamin A, iron, vitamin D, iodine, zinc, vitamin B12), iron-folic acid (IFA) supplementation, multiple micronutrient (MMN) supplementation, and lipid-based nutrient supplementation (LNS) during pregnancy. Search Methods We searched papers published from 1995 to 31 October 2019 (related programmes and good quality studies pre-1995 were limited) in CAB Abstracts, CINAHL, Cochrane Central Register of Controlled Trials, Embase, International Initiative for Impact Evaluations, LILACS, Medline, POPLINE, Web of Science, WHOLIS, ProQuest Dissertations & Theses Global, R4D, WHO International Clinical Trials Registry Platform. Non-indexed grey literature searches were conducted using Google, Google Scholar, and web pages of key international nutrition agencies. Selection Criteria We included randomized controlled trials (individual and cluster-randomized) and quasi-experimental studies that evaluated micronutrient supplementation in healthy, pregnant women of any age and parity living in a LMIC. LMIC were defined by the World Bank Group at the time of the search for this review. While the aim was to include healthy pregnant women, it is likely that these populations had one or more micronutrient deficiencies at baseline; women were not excluded on this basis. Data Collection and Analysis Two authors independently assessed studies for inclusion and risk of bias, and conducted data extraction. Data were matched to check for accuracy. Quality of evidence was assessed using the GRADE approach. Main Results A total of 314 papers across 72 studies (451,723 women) were eligible for inclusion, of which 64 studies (439,649 women) contributed to meta-analyses. Seven studies assessed iron-folic acid (IFA) supplementation versus folic acid; 34 studies assessed MMN vs. IFA; 4 studies assessed LNS vs. MMN; 13 evaluated iron; 13 assessed zinc; 9 evaluated vitamin A; 11 assessed vitamin D; and 6 assessed calcium. Several studies were eligible for inclusion in multiple types of supplementation. IFA compared to folic acid showed a large and significant (48%) reduction in the risk of maternal anaemia (average risk ratio (RR) 0.52, 95% CI 0.41 to 0.66; studies = 5; participants = 15,540; moderate-quality evidence). As well, IFA supplementation demonstrated a smaller but significant, 12% reduction in risk of low birthweight (LBW) babies (average RR 0.88, 95% CI 0.78 to 0.99; studies = 4; participants = 17,257; high-quality evidence). MMN supplementation was defined as any supplement that contained at least 3 micronutrients. Post-hoc analyses were conducted, where possible, comparing the differences in effect of MMN with 4+ components and MMN with 3 or 4 components. When compared to iron with or without FA, MMN supplementation reduced the risk of LBW by 15% (average RR 0.85, 95% CI 0.77 to 0.93; studies = 28; participants = 79,972); this effect was greater in MMN with >4 micronutrients (average RR 0.79, 95% CI 0.71 to 0.88; studies = 19; participants = 68,138 versus average RR 1.01, 95% CI 0.92 to 1.11; studies = 9; participants = 11,834). There was a small and significant reduction in the risk of stillbirths (average RR 0.91; 95% CI 0.86 to 0.98; studies = 22; participants = 96,772) and a small and significant effect on the risk of small-for-gestational age (SGA) (average RR 0.93; 95% CI 0.88 to 0.98; studies = 19; participants = 52,965). For stillbirths and SGA, the effects were greater among those provided MMN with 4+ micronutrients. Children whose mothers had been supplemented with MMN, compared to IFA, demonstrated a 16% reduced risk of diarrhea (average RR 0.84; 95% CI 0.76 to 0.92; studies = 4; participants = 3,142). LNS supplementation, compared to MMN, made no difference to any outcome; however, the evidence is limited. Iron supplementation, when compared to no iron or placebo, showed a large and significant effect on maternal anaemia, a reduction of 47% (average RR 0.53, 95% CI 0.43 to 0.65; studies = 6; participants = 15,737; moderate-quality evidence) and a small and significant effect on LBW (average RR 0.88, 95% CI 0.78 to 0.99; studies = 4; participants = 17,257; high-quality evidence). Zinc and vitamin A supplementation, each both compared to placebo, had no impact on any outcome examined with the exception of potentially improving serum/plasma zinc (mean difference (MD) 0.43 umol/L; 95% CI -0.04 to 0.89; studies = 5; participants = 1,202) and serum/plasma retinol (MD 0.13 umol/L; 95% CI -0.03 to 0.30; studies = 6; participants = 1,654), respectively. When compared to placebo, vitamin D supplementation may have reduced the risk of preterm births (average RR 0.64; 95% CI 0.40 to 1.04; studies = 7; participants = 1,262), though the upper CI just crosses the line of no effect. Similarly, calcium supplementation versus placebo may have improved rates of pre-eclampsia/eclampsia (average RR 0.45; 95% CI 0.19 to 1.06; studies = 4; participants = 9,616), though the upper CI just crosses 1. Authors' Conclusions The findings suggest that MMN and vitamin supplementation improve maternal and child health outcomes, including maternal anaemia, LBW, preterm birth, SGA, stillbirths, micronutrient deficiencies, and morbidities, including pre-eclampsia/eclampsia and diarrhea among children. MMN supplementation demonstrated a beneficial impact on the most number of outcomes. In addition, MMN with >4 micronutrients appeared to be more impactful than MMN with only 3 or 4 micronutrients included in the tablet. Very few studies conducted longitudinal analysis on longer-term health outcomes for the child, such as anthropometric measures and developmental outcomes; this may be an important area for future research. This review may provide some basis to guide continual discourse around replacing IFA supplementation with MMN along with the use of single micronutrient supplementation programs for specific outcomes.
Collapse
Affiliation(s)
- Emily C. Keats
- Centre for Global Child HealthThe Hospital for Sick ChildrenTorontoCanada
| | - Christina Oh
- Centre for Global Child HealthThe Hospital for Sick ChildrenTorontoCanada
| | - Tamara Chau
- Centre for Global Child HealthThe Hospital for Sick ChildrenTorontoCanada
| | - Dina S. Khalifa
- Centre for Global Child HealthThe Hospital for Sick ChildrenTorontoCanada
| | - Aamer Imdad
- PediatricsUpstate Medical University, SyracuseNew YorkUSA
| | - Zulfiqar A. Bhutta
- Centre for Global Child HealthThe Hospital for Sick ChildrenTorontoCanada
| |
Collapse
|
30
|
Camm EJ, Cross CM, Kane AD, Tarry-Adkins JL, Ozanne SE, Giussani DA. Maternal antioxidant treatment protects adult offspring against memory loss and hippocampal atrophy in a rodent model of developmental hypoxia. FASEB J 2021; 35:e21477. [PMID: 33891326 DOI: 10.1096/fj.202002557rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/11/2021] [Accepted: 02/11/2021] [Indexed: 02/02/2023]
Abstract
Chronic fetal hypoxia is one of the most common outcomes in complicated pregnancy in humans. Despite this, its effects on the long-term health of the brain in offspring are largely unknown. Here, we investigated in rats whether hypoxic pregnancy affects brain structure and function in the adult offspring and explored underlying mechanisms with maternal antioxidant intervention. Pregnant rats were randomly chosen for normoxic or hypoxic (13% oxygen) pregnancy with or without maternal supplementation with vitamin C in their drinking water. In one cohort, the placenta and fetal tissues were collected at the end of gestation. In another, dams were allowed to deliver naturally, and offspring were reared under normoxic conditions until 4 months of age (young adult). Between 3.5 and 4 months, the behavior, cognition and brains of the adult offspring were studied. We demonstrated that prenatal hypoxia reduced neuronal number, as well as vascular and synaptic density, in the hippocampus, significantly impairing memory function in the adult offspring. These adverse effects of prenatal hypoxia were independent of the hypoxic pregnancy inducing fetal growth restriction or elevations in maternal or fetal plasma glucocorticoid levels. Maternal vitamin C supplementation during hypoxic pregnancy protected against oxidative stress in the placenta and prevented the adverse effects of prenatal hypoxia on hippocampal atrophy and memory loss in the adult offspring. Therefore, these data provide a link between prenatal hypoxia, placental oxidative stress, and offspring brain health in later life, providing insight into mechanism and identifying a therapeutic strategy.
Collapse
Affiliation(s)
- Emily J Camm
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - Christine M Cross
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - Andrew D Kane
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - Jane L Tarry-Adkins
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.,Cambridge Strategic Initiative in Reproduction, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK.,Cambridge Strategic Initiative in Reproduction, Cambridge, UK
| |
Collapse
|
31
|
Roberts JM, Rich-Edwards JW, McElrath TF, Garmire L, Myatt L. Subtypes of Preeclampsia: Recognition and Determining Clinical Usefulness. Hypertension 2021; 77:1430-1441. [PMID: 33775113 DOI: 10.1161/hypertensionaha.120.14781] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The concept that preeclampsia is a multisystemic syndrome is appreciated in both research and clinical care. Our understanding of pathophysiology recognizes the role of inflammation, oxidative and endoplasm reticulum stress, and angiogenic dysfunction. Yet, we have not progressed greatly toward clinically useful prediction nor had substantial success in prevention or treatment. One possibility is that the maternal syndrome may be reached through different pathophysiological pathways, that is, subtypes of preeclampsia, that in their specificity yield more clinical utility. For example, early and late onset preeclampsia are increasingly acknowledged as different pathophysiological processes leading to a common presentation. Other subtypes of preeclampsia are supported by disparate clinical outcomes, long-range prognosis, organ systems involved, and risk factors. These insights have been supplemented by discovery-driven methods, which cluster preeclampsia cases into groups indicating different pathophysiologies. In this presentation, we review likely subtypes based on current knowledge and suggest others. We present a consideration of the requirements for a clinically meaningful preeclampsia subtype. A useful subtype should (1) identify a specific pathophysiological pathway or (2) specifically indicate maternal or fetal outcome, (3) be recognizable in a clinically useful time frame, and (4) these results should be reproducible and generalizable (but at varying frequency) including in low resource settings. We recommend that the default consideration be that preeclampsia includes several subtypes rather than trying to force all cases into a single pathophysiological pathway. The recognition of subtypes and deciphering their different pathophysiologies will provide specific targets for prevention, prediction, and treatment directing personalized care.
Collapse
Affiliation(s)
- James M Roberts
- Magee-Womens Research Institute, Department of Obstetrics Gynecology and Reproductive Sciences, Epidemiology and Clinical and Translational Research, University of Pittsburgh (J.M.R.)
| | - Janet W Rich-Edwards
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA (J.W.R.-E.).,Division of Women's Health, Department of Medicine (J.W.R.-E.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Thomas F McElrath
- Division of Maternal-Fetal Medicine (T.F.M.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Lana Garmire
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan (L.G.)
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Moore Institute of Nutrition and Wellness, Oregon Health and Science University (L.M.)
| | | |
Collapse
|
32
|
Liu Y, Li N, Mei Z, Li Z, Ye R, Zhang L, Li H, Zhang Y, Liu JM, Serdula MK. Effects of prenatal micronutrients supplementation timing on pregnancy-induced hypertension: Secondary analysis of a double-blind randomized controlled trial. MATERNAL AND CHILD NUTRITION 2021; 17:e13157. [PMID: 33594802 PMCID: PMC8189207 DOI: 10.1111/mcn.13157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 01/21/2021] [Accepted: 01/24/2021] [Indexed: 11/27/2022]
Abstract
In this secondary analysis of data from a double‐blind randomized controlled trial (clinicaltrials.gov identifier: NCT00133744) of micronutrient supplementation (multiple micronutrients [MMN], iron–folic acid [IFA] and folic acid [FA] alone), we examined the potential modifying effect of gestational age at enrolment on the association of antenatal supplementation and pregnancy‐induced hypertension (PIH). We included 18,775 nulliparous pregnant women with mild or no anaemia who were enrolled at 20 weeks of gestation or earlier from five counties of northern China. Women were randomly assigned to receive daily FA, IFA or MMN from enrolment until delivery. We used logistic regression to evaluate the association between PIH and timing of micronutrient supplementation. The incidence of PIH was statistically significantly lower among women who began MMN supplementation before 12 gestational weeks compared with women who began MMN supplementation at 12 weeks or later (RR = 0.74, 95% CI: 0.60–0.91). A similar protective effect was observed for both early‐onset (<28 weeks, RR 0.45, 0.21–0.96) and late‐onset of PIH (≥28 weeks, RR 0.77, 0.63–0.96). No statistically significant association was observed between PIH occurrence and timing of supplementation for FA or IFA. Maternal MMN supplementation and antenatal enrolment during the first trimester of pregnancy appeared to be of importance in preventing both early‐ and late‐onset of PIH.
Collapse
Affiliation(s)
- Yingying Liu
- Institute of Reproductive and Child Health/Ministry of Health Key Laboratory of Reproductive Health and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Nan Li
- Institute of Reproductive and Child Health/Ministry of Health Key Laboratory of Reproductive Health and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Zuguo Mei
- Division of Nutrition, Physical Activity, and Obesity, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Zhiwen Li
- Institute of Reproductive and Child Health/Ministry of Health Key Laboratory of Reproductive Health and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Rongwei Ye
- Institute of Reproductive and Child Health/Ministry of Health Key Laboratory of Reproductive Health and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Le Zhang
- Institute of Reproductive and Child Health/Ministry of Health Key Laboratory of Reproductive Health and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Hongtian Li
- Institute of Reproductive and Child Health/Ministry of Health Key Laboratory of Reproductive Health and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Yali Zhang
- Institute of Reproductive and Child Health/Ministry of Health Key Laboratory of Reproductive Health and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Jian-Meng Liu
- Institute of Reproductive and Child Health/Ministry of Health Key Laboratory of Reproductive Health and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Mary K Serdula
- Division of Nutrition, Physical Activity, and Obesity, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
33
|
Paffoni A, Reschini M, Noli SA, Viganò P, Parazzini F, Somigliana E. Folate Levels and Pregnancy Rate in Women Undergoing Assisted Reproductive Techniques: a Systematic Review and Meta-analysis. Reprod Sci 2021; 29:341-356. [DOI: 10.1007/s43032-021-00467-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 01/11/2021] [Indexed: 12/29/2022]
|
34
|
Kinshella MLW, Omar S, Scherbinsky K, Vidler M, Magee LA, von Dadelszen P, Moore SE, Elango R, The PRECISE Conceptual Framework Working Group. Effects of Maternal Nutritional Supplements and Dietary Interventions on Placental Complications: An Umbrella Review, Meta-Analysis and Evidence Map. Nutrients 2021; 13:472. [PMID: 33573262 PMCID: PMC7912620 DOI: 10.3390/nu13020472] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 01/26/2021] [Indexed: 01/08/2023] Open
Abstract
The placenta is a vital, multi-functional organ that acts as an interface between maternal and fetal circulation during pregnancy. Nutritional deficiencies during pregnancy alter placental development and function, leading to adverse pregnancy outcomes, such as pre-eclampsia, infants with small for gestational age and low birthweight, preterm birth, stillbirths and maternal mortality. Maternal nutritional supplementation may help to mitigate the risks, but the evidence base is difficult to navigate. The primary purpose of this umbrella review is to map the evidence on the effects of maternal nutritional supplements and dietary interventions on pregnancy outcomes related to placental disorders and maternal mortality. A systematic search was performed on seven electronic databases, the PROSPERO register and references lists of identified papers. The results were screened in a three-stage process based on title, abstract and full-text by two independent reviewers. Randomized controlled trial meta-analyses on the efficacy of maternal nutritional supplements or dietary interventions were included. There were 91 meta-analyses included, covering 23 types of supplements and three types of dietary interventions. We found evidence that supports supplementary vitamin D and/or calcium, omega-3, multiple micronutrients, lipid-based nutrients, and balanced protein energy in reducing the risks of adverse maternal and fetal health outcomes. However, these findings are limited by poor quality of evidence. Nutrient combinations show promise and support a paradigm shift to maternal dietary balance, rather than single micronutrient deficiencies, to improve maternal and fetal health. The review is registered at PROSPERO (CRD42020160887).
Collapse
Affiliation(s)
- Mai-Lei Woo Kinshella
- Department of Obstetrics and Gynaecology, BC Children’s and Women’s Hospital and University of British Columbia, Vancouver, BC V6Z 2K8, Canada; (M.-L.W.K.); (S.O.); (K.S.); (M.V.); (L.A.M.); (P.v.D.)
| | - Shazmeen Omar
- Department of Obstetrics and Gynaecology, BC Children’s and Women’s Hospital and University of British Columbia, Vancouver, BC V6Z 2K8, Canada; (M.-L.W.K.); (S.O.); (K.S.); (M.V.); (L.A.M.); (P.v.D.)
| | - Kerri Scherbinsky
- Department of Obstetrics and Gynaecology, BC Children’s and Women’s Hospital and University of British Columbia, Vancouver, BC V6Z 2K8, Canada; (M.-L.W.K.); (S.O.); (K.S.); (M.V.); (L.A.M.); (P.v.D.)
- Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 0B3, Canada
| | - Marianne Vidler
- Department of Obstetrics and Gynaecology, BC Children’s and Women’s Hospital and University of British Columbia, Vancouver, BC V6Z 2K8, Canada; (M.-L.W.K.); (S.O.); (K.S.); (M.V.); (L.A.M.); (P.v.D.)
| | - Laura A. Magee
- Department of Obstetrics and Gynaecology, BC Children’s and Women’s Hospital and University of British Columbia, Vancouver, BC V6Z 2K8, Canada; (M.-L.W.K.); (S.O.); (K.S.); (M.V.); (L.A.M.); (P.v.D.)
- Department of Women & Children’s Health, King’s College London, London WC2R 2LS, UK;
| | - Peter von Dadelszen
- Department of Obstetrics and Gynaecology, BC Children’s and Women’s Hospital and University of British Columbia, Vancouver, BC V6Z 2K8, Canada; (M.-L.W.K.); (S.O.); (K.S.); (M.V.); (L.A.M.); (P.v.D.)
- Department of Women & Children’s Health, King’s College London, London WC2R 2LS, UK;
| | - Sophie E. Moore
- Department of Women & Children’s Health, King’s College London, London WC2R 2LS, UK;
- Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, P.O. Box 273 Banjul, The Gambia
| | - Rajavel Elango
- Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 0B3, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Division of Neonatology, BC Women’s Hospital and Health Centre, Vancouver, BC V6H 3N1, Canada
| | | |
Collapse
|
35
|
Aminuddin NA, Sutan R, Mahdy ZA. Role of Palm Oil Vitamin E in Preventing Pre-eclampsia: A Secondary Analysis of a Randomized Clinical Trial Following ISSHP Reclassification. Front Med (Lausanne) 2021; 7:596405. [PMID: 33553199 PMCID: PMC7859347 DOI: 10.3389/fmed.2020.596405] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/30/2020] [Indexed: 11/17/2022] Open
Abstract
Background: Preeclampsia is a significant cause of maternal and perinatal mortality worldwide. Oxidative stress plays a key role in its pathophysiology, hence antioxidants such as tocotrienol may be preventive against preeclampsia. In 2018, the ISSHP revised the definition of preeclampsia. In accordance with the new definition, we report a secondary data analysis from a clinical trial comparing palm oil vitamin E in the form of tocotrienol-rich fraction (TRF) against placebo, in preventing preeclampsia. Method: A randomized double-blind controlled trial was conducted in 2002–2005 to assess the benefits of TRF in preeclampsia prevention. A total of 299 primigravidae were recruited. The intervention group was supplemented with TRF 100 mg daily in super-olein capsules, whereas the placebo group was prescribed super-olein capsules without TRF, beginning from 12 to 16 gestational weeks until delivery. The primary outcome measure was incidence of preeclampsia. Results: The total incidence of pregnancy induced hypertension (PIH) was 5%, whereas the incidence of preeclampsia was 2.3%. The odds of developing PIH (adjusted OR 0.254; 95% CI: 0.07–0.93; p–value 0.038) and preeclampsia (adjusted OR 0.030; 95% CI: 0.001–0.65; p-value 0.025) were significantly lower in the TRF arm compared to the placebo arm. Conclusion: Antenatal supplementation with palm oil vitamin E in the form of TRF is associated with significant reductions in the incidence of preeclampsia and PIH in a single urban tertiary hospital. Palm oil vitamin E deserves further scrutiny as a potential public health preventive measure against preeclampsia and PIH.
Collapse
Affiliation(s)
- Nurul Afzan Aminuddin
- Community Health Department, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| | - Rosnah Sutan
- Community Health Department, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| | - Zaleha Abdullah Mahdy
- Obstetrics and Gynaecology Department, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| |
Collapse
|
36
|
Korenc M, Zieleskiewicz L, Stopar Pintaric T, Blajic I, Ambrozic J, Lucovnik M. The effect of vitamin C on pulmonary oedema in patients with severe preeclampsia: A single-centre, randomised, placebo-controlled, double-blind trial. Anaesth Crit Care Pain Med 2021; 40:100800. [PMID: 33453456 DOI: 10.1016/j.accpm.2021.100800] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/18/2020] [Accepted: 09/22/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To determine whether vitamin C in the first three days postpartum reduces pulmonary oedema (PE) assessed by lung ultrasound in patients with severe preeclampsia. DESIGN Randomised, placebo-controlled, double-blind trial. SETTING Tertiary perinatal centre. POPULATION Consecutively admitted patients with singleton pregnancies complicated by severe preeclampsia. METHODS Thirty-four patients received vitamin C (1.5 g/6 h) (n = 17) or placebo (n = 17) at days 1, 2, and 3 postdelivery. Mann-Whitney-U test was used to compare vitamin C vs placebo groups. A p ≤ 0.05 was considered statistically significant. MAIN OUTCOME MEASURES Lung ultrasound was performed once daily in the first three days following delivery. Echo Comet Score (ECS) on day 1 postdelivery was the primary outcome studied and was obtained using the 28-rib interspaces technique. ECS on days 2 and 3 postdelivery were secondary outcomes. RESULTS There was no significant difference in ECS on day 1 (median 23 (inter-quartile range (IQR) 21-61) vs 18 (IQR 8-35); p = 0.31). All ultrasound examinations on day 1 were performed within six hours from delivery. On days 2 and 3, ECS was significantly lower in vitamin C group compared to placebo (8 (IQR 3-14) vs 35 (IQR 15-78); p = 0.03 and 5 (IQR 3-10) vs 18 (IQR 18-44); p = 0.04, respectively). CONCLUSION A single dose of intravenous vitamin C did not reduce PE in postpartum patients with severe preeclampsia on day 1 after delivery. Repeated doses, however, seem to have a delayed effect with a reduction in PE detected on ultrasound on days 2 and 3 following delivery. TRIAL REGISTRATION This trial is registered at ClinicalTrials.gov: ID NCT03451266 (https://clinicaltrials.gov/ct2/show/NCT03451266?term=NCT03451266&draw=2&rank=1).
Collapse
Affiliation(s)
- Monika Korenc
- Department of Perinatology, Division of Obstetrics and Gynaecology, University Medical Centre Ljubljana, Slovenia
| | - Laurent Zieleskiewicz
- Department of Anaesthesiology and Critical Care Medicine, North Hospital, Aix Marseille University, Marseille, France
| | - Tatjana Stopar Pintaric
- Department of Anaesthesiology and Intensive Therapy, University Medical Centre Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Slovenia
| | - Iva Blajic
- Department of Anaesthesiology and Intensive Therapy, University Medical Centre Ljubljana, Slovenia
| | - Jana Ambrozic
- Department of Cardiology, University Medical Centre Ljubljana, Slovenia
| | - Miha Lucovnik
- Department of Perinatology, Division of Obstetrics and Gynaecology, University Medical Centre Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Slovenia.
| |
Collapse
|
37
|
Hinkle SN, Zhang C, Grantz KL, Sciscione A, Wing DA, Grobman WA, Newman RB, D'Alton ME, Skupski D, Nageotte MP, Ranzini AC, Owen J, Chein EK, Craigo S, Yisahak SF, Liu A, Albert PS, Louis GMB, Grewal J. Nutrition during Pregnancy: Findings from the National Institute of Child Health and Human Development (NICHD) Fetal Growth Studies-Singleton Cohort. Curr Dev Nutr 2021; 5:nzaa182. [PMID: 33553996 PMCID: PMC7846139 DOI: 10.1093/cdn/nzaa182] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Accumulating evidence indicates that maternal diets are important for optimizing maternal and offspring health. Existing research lacks comprehensive profiles of maternal diets throughout pregnancy, especially in a racially/ethnically diverse obstetrical population. OBJECTIVE The aim was to characterize diets in a longitudinal US pregnancy cohort by trimester, race/ethnicity, and prepregnancy BMI. METHODS Data were obtained from pregnant women in the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) Fetal Growth Studies-Singleton cohort (2009-2013). A food-frequency questionnaire (FFQ) at 8-13 wk of gestation assessed periconception and first-trimester diet (n = 1615). Automated, self-administered, 24-h dietary recalls targeted at 16-22, 24-29, 30-33, and 34-37 wk of gestation assessed second- and third-trimester diets (n = 1817 women/6791 recalls). The Healthy Eating Index-2010 (HEI-2010) assessed diet quality (i.e., adherence to US Dietary Guidelines). Variations in weighted energy-adjusted means for foods and nutrients were examined by trimester, self-identified race/ethnicity, and self-reported prepregnancy BMI. RESULTS Mean (95% CI) HEI-2010 was 65.9 (64.9, 67.0) during periconception to the first trimester assessed with an FFQ and 51.6 (50.8, 52.4) and 51.5 (50.7, 52.3) during the second trimester and third trimester, respectively, assessed using 24-h recalls. No significant differences were observed between the second and third trimester in macronutrients, micronutrients, foods, or HEI-2010 components (P ≥ 0.05). Periconception to first-trimester HEI-2010 was highest among Asian/Pacific Islander [67.2 (65.9, 68.6)] and lowest among non-Hispanic Black [58.7 (57.5, 60.0)] women and highest among women with normal weight [67.2 (66.1, 68.4)] and lowest among women with obesity [63.5 (62.1, 64.9)]. Similar rankings were observed in the second/third trimesters. CONCLUSIONS Most pregnant women in this cohort reported dietary intakes that, on average, did not meet US Dietary Guidelines for nonpregnant individuals. Also, diet differed across race/ethnic groups and by prepregnancy BMI, with the lowest overall dietary quality in all trimesters among non-Hispanic Black women and women with obesity. No meaningful changes in dietary intake were observed between the second and third trimesters.
Collapse
Affiliation(s)
- Stefanie N Hinkle
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Cuilin Zhang
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Katherine L Grantz
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Anthony Sciscione
- Department of Obstetrics and Gynecology, Christiana Care Health System, Newark, DE, USA
| | - Deborah A Wing
- Korn Ferry, Los Angeles, CA, USA
- Division of Maternal-Fetal Medicine, University of California, Irvine, CA, USA
| | - William A Grobman
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Roger B Newman
- Department of Obstetrics and Gynecology, Medical University of South Carolina, Charleston, SC, USA
| | - Mary E D'Alton
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, USA
| | - Daniel Skupski
- Department of Obstetrics and Gynecology, New York Hospital Queens, Queens, NY, USA
| | - Michael P Nageotte
- Department of Obstetrics and Gynecology, Miller Children's Hospital/Long Beach Memorial Medical Center, Long Beach, CA, USA
| | - Angela C Ranzini
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, St Peter's University Hospital, New Brunswick, NJ, USA
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, MetroHealth Medical Center/Case Western Reserve University, Cleveland, OH, USA
| | - John Owen
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology University of Alabama, Birmingham, School of Medicine, Birmingham, AL, USA
| | - Edward K Chein
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Women and Infants Hospital of Rhode Island, Providence, RI, USA
| | - Sabrina Craigo
- Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, MA, USA
| | - Samrawit F Yisahak
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Aiyi Liu
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Paul S Albert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Germaine M Buck Louis
- Dean's Office, College of Health and Human Services, George Mason University, Fairfax, VA, USA
| | - Jagteshwar Grewal
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| |
Collapse
|
38
|
Snell KIE, Allotey J, Smuk M, Hooper R, Chan C, Ahmed A, Chappell LC, Von Dadelszen P, Green M, Kenny L, Khalil A, Khan KS, Mol BW, Myers J, Poston L, Thilaganathan B, Staff AC, Smith GCS, Ganzevoort W, Laivuori H, Odibo AO, Arenas Ramírez J, Kingdom J, Daskalakis G, Farrar D, Baschat AA, Seed PT, Prefumo F, da Silva Costa F, Groen H, Audibert F, Masse J, Skråstad RB, Salvesen KÅ, Haavaldsen C, Nagata C, Rumbold AR, Heinonen S, Askie LM, Smits LJM, Vinter CA, Magnus P, Eero K, Villa PM, Jenum AK, Andersen LB, Norman JE, Ohkuchi A, Eskild A, Bhattacharya S, McAuliffe FM, Galindo A, Herraiz I, Carbillon L, Klipstein-Grobusch K, Yeo SA, Browne JL, Moons KGM, Riley RD, Thangaratinam S. External validation of prognostic models predicting pre-eclampsia: individual participant data meta-analysis. BMC Med 2020; 18:302. [PMID: 33131506 PMCID: PMC7604970 DOI: 10.1186/s12916-020-01766-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/26/2020] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Pre-eclampsia is a leading cause of maternal and perinatal mortality and morbidity. Early identification of women at risk during pregnancy is required to plan management. Although there are many published prediction models for pre-eclampsia, few have been validated in external data. Our objective was to externally validate published prediction models for pre-eclampsia using individual participant data (IPD) from UK studies, to evaluate whether any of the models can accurately predict the condition when used within the UK healthcare setting. METHODS IPD from 11 UK cohort studies (217,415 pregnant women) within the International Prediction of Pregnancy Complications (IPPIC) pre-eclampsia network contributed to external validation of published prediction models, identified by systematic review. Cohorts that measured all predictor variables in at least one of the identified models and reported pre-eclampsia as an outcome were included for validation. We reported the model predictive performance as discrimination (C-statistic), calibration (calibration plots, calibration slope, calibration-in-the-large), and net benefit. Performance measures were estimated separately in each available study and then, where possible, combined across studies in a random-effects meta-analysis. RESULTS Of 131 published models, 67 provided the full model equation and 24 could be validated in 11 UK cohorts. Most of the models showed modest discrimination with summary C-statistics between 0.6 and 0.7. The calibration of the predicted compared to observed risk was generally poor for most models with observed calibration slopes less than 1, indicating that predictions were generally too extreme, although confidence intervals were wide. There was large between-study heterogeneity in each model's calibration-in-the-large, suggesting poor calibration of the predicted overall risk across populations. In a subset of models, the net benefit of using the models to inform clinical decisions appeared small and limited to probability thresholds between 5 and 7%. CONCLUSIONS The evaluated models had modest predictive performance, with key limitations such as poor calibration (likely due to overfitting in the original development datasets), substantial heterogeneity, and small net benefit across settings. The evidence to support the use of these prediction models for pre-eclampsia in clinical decision-making is limited. Any models that we could not validate should be examined in terms of their predictive performance, net benefit, and heterogeneity across multiple UK settings before consideration for use in practice. TRIAL REGISTRATION PROSPERO ID: CRD42015029349 .
Collapse
Affiliation(s)
- Kym I E Snell
- Centre for Prognosis Research, School of Primary, Community and Social Care, Keele University, Keele, UK.
| | - John Allotey
- Barts Research Centre for Women's Health (BARC), Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Pragmatic Clinical Trials Unit, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Melanie Smuk
- Pragmatic Clinical Trials Unit, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Richard Hooper
- Pragmatic Clinical Trials Unit, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Claire Chan
- Pragmatic Clinical Trials Unit, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Asif Ahmed
- MirZyme Therapeutics, Innovation Birmingham Campus, Birmingham, UK
| | - Lucy C Chappell
- Department of Women and Children's Health, School of Life Course Sciences, King's College London, London, UK
| | - Peter Von Dadelszen
- Department of Women and Children's Health, School of Life Course Sciences, King's College London, London, UK
| | - Marcus Green
- Action on Pre-eclampsia (APEC) Charity, Worcestershire, UK
| | - Louise Kenny
- Faculty Health & Life Sciences, University of Liverpool, Liverpool, UK
| | - Asma Khalil
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust and Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Khalid S Khan
- Barts Research Centre for Women's Health (BARC), Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Pragmatic Clinical Trials Unit, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ben W Mol
- Department of Obstetrics and Gynaecology, Monash University, Monash Medical Centre, Clayton, Victoria, Australia
| | - Jenny Myers
- Maternal and Fetal Health Research Centre, Manchester Academic Health Science Centre, University of Manchester, Central Manchester NHS Trust, Manchester, UK
| | - Lucilla Poston
- Department of Women and Children's Health, School of Life Course Sciences, King's College London, London, UK
| | - Basky Thilaganathan
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust and Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Anne C Staff
- Division of Obstetrics and Gynaecology, Oslo University Hospital, and Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Gordon C S Smith
- Department of Obstetrics and Gynaecology, NIHR Biomedical Research Centre, Cambridge University, Cambridge, UK
| | - Wessel Ganzevoort
- Department of Obstetrics, Amsterdam UMC University of Amsterdam, Amsterdam, The Netherlands
| | - Hannele Laivuori
- Department of Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Department of Obstetrics and Gynecology, Faculty of Medicine and Health Technology, Tampere University Hospital and Tampere University, Tampere, Finland
| | | | - Javier Arenas Ramírez
- Department of Obstetrics and Gynaecology, University Hospital de Cabueñes, Gijón, Spain
| | - John Kingdom
- Maternal-Fetal Medicine Division, Department OBGYN, Mount Sinai Hospital, University of Toronto, Toronto, Canada
| | - George Daskalakis
- Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Alexandra Hospital, Athens, Greece
| | - Diane Farrar
- Bradford Institute for Health Research, Bradford Teaching Hospitals, Bradford, UK
| | - Ahmet A Baschat
- Johns Hopkins Center for Fetal Therapy, Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul T Seed
- Department of Women and Children's Health, School of Life Course Sciences, King's College London, London, UK
| | - Federico Prefumo
- Department of Obstetrics and Gynaecology, University of Brescia, Brescia, Italy
| | - Fabricio da Silva Costa
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Henk Groen
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Francois Audibert
- Department of Obstetrics and Gynecology, CHU Ste Justine, Université de Montréal, Montreal, Canada
| | - Jacques Masse
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Quebec City, Canada
| | - Ragnhild B Skråstad
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
- Department of Clinical Pharmacology, St. Olav University Hospital, Trondheim, Norway
| | - Kjell Å Salvesen
- Department of Obstetrics and Gynecology, Trondheim University Hospital, Trondheim, Norway
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Camilla Haavaldsen
- Department of Obstetrics and Gynaecology, Akershus University Hospital, Lørenskog, Norway
| | - Chie Nagata
- Department of Education for Clinical Research, National Center for Child Health and Development, Tokyo, Japan
| | - Alice R Rumbold
- South Australian Health and Medical Research Institute and Robinson Research Institute, The University of Adelaide, Adelaide, Australia
| | - Seppo Heinonen
- Department of Obstetrics and Gynaecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Lisa M Askie
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Luc J M Smits
- Care and Public Health Research Institute, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Christina A Vinter
- Department of Gynecology and Obstetrics, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Kajantie Eero
- National Institute for Health and Welfare, Helsinki, Finland
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pia M Villa
- Department of Obstetrics and Gynaecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anne K Jenum
- General Practice Research Unit (AFE), Department of General Practice, Institute of Health and Society, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Louise B Andersen
- Institute for Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Obstetrics and Gynecology, Odense University Hospital, Odense, Denmark
| | - Jane E Norman
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Akihide Ohkuchi
- Department of Obstetrics and Gynecology, Jichi Medical University School of Medicine, Shimotsuke-shi, Tochigi, Japan
| | - Anne Eskild
- Department of Obstetrics and Gynaecology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Sohinee Bhattacharya
- Obstetrics & Gynaecology, Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Fionnuala M McAuliffe
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Ireland
| | - Alberto Galindo
- Fetal Medicine Unit, Maternal and Child Health and Development Network (SAMID), Department of Obstetrics and Gynaecology, Hospital Universitario, Instituto de Investigación Hospital, Universidad Complutense de Madrid, Madrid, Spain
| | - Ignacio Herraiz
- Fetal Medicine Unit, Maternal and Child Health and Development Network (SAMID), Department of Obstetrics and Gynaecology, Hospital Universitario, Instituto de Investigación Hospital, Universidad Complutense de Madrid, Madrid, Spain
| | - Lionel Carbillon
- Department of Obstetrics and Gynecology, Assistance Publique-Hôpitaux de Paris Université Paris, Paris, France
| | - Kerstin Klipstein-Grobusch
- Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Seon Ae Yeo
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joyce L Browne
- Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Karel G M Moons
- Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
- Cochrane Netherlands, Utrecht, The Netherlands
| | - Richard D Riley
- Centre for Prognosis Research, School of Primary, Community and Social Care, Keele University, Keele, UK
| | - Shakila Thangaratinam
- Institute of Metabolism and Systems Research, WHO Collaborating Centre for Women's Health, University of Birmingham, Birmingham, UK
| |
Collapse
|
39
|
NAWSHERWAN, KHAN S, ZEB F, SHOAIB M, NABI G, UL HAQ I, XU K, LI H. Selected Micronutrients: An Option to Boost Immunity against COVID-19 and Prevent Adverse Pregnancy Outcomes in Pregnant Women: A Narrative Review. IRANIAN JOURNAL OF PUBLIC HEALTH 2020; 49:2032-2043. [PMID: 33708724 PMCID: PMC7917498 DOI: 10.18502/ijph.v49i11.4717] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 08/19/2020] [Indexed: 12/17/2022]
Abstract
The coronavirus disease-19 (COVID-19) negatively affects immune system. It is linked with adverse pregnancy outcomes. These complications may be linked with the infections mediated deficiency of micronutrients in pregnant women. COVID-19 cause's malabsorption of micronutrients thereby increases the risk of their deficiency. Both micronutrients deficiencies and poor micronutrients intake can compromise immune function and may increase the risk of pregnancy complications associated with COVID-19 infection. Vita-min A, C, D, E, and selected minerals iron (Fe), selenium (Se), and zinc (Zn) are the micronutrients essential for immuno-competency and play a significant role in the prevention of adverse pregnancy outcomes. Immune function and pregnancy outcomes can be improved by adequate intake of micronutrients in diet or in supplements form. Based on regulatory links between viral infection, micronutrients, immunity, and pregnancy outcomes, this review highlights the role of micronutrients in boosting immunity to reduce or prevent pregnancy complications in COVID-19 infected women.
Collapse
Affiliation(s)
- NAWSHERWAN
- Department of Preventive Medicine, School of Health Sciences, Wuhan University, Wuhan, Hubei, China
| | - Suliman KHAN
- Department of Cerebrovascular Diseases, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Falak ZEB
- Department of Nutrition and Dietetics, National University of Medical Sciences, Islamabad, Pakistan
| | - Muhammad SHOAIB
- Department of Chemistry, Government of Postgraduate College Samanabad, Faisalabad, Pakistan
| | - Ghulam NABI
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Ijaz UL HAQ
- Department of Public Health and Nutrition, The University of Haripur, Khyber Pakhtunkhwa, Pakistan
| | - Kang XU
- Department of Medicine, Taixing People Hospital, Taizhou, Jiangsu, China
| | - Hui LI
- Department of Medicine, Taixing People Hospital, Taizhou, Jiangsu, China
| |
Collapse
|
40
|
Abdi N, Rozrokh A, Alavi A, Zare S, Vafaei H, Asadi N, Kasraeian M, Hessami K. The effect of aspirin on preeclampsia, intrauterine growth restriction and preterm delivery among healthy pregnancies with a history of preeclampsia. J Chin Med Assoc 2020; 83:852-857. [PMID: 32773581 PMCID: PMC7478204 DOI: 10.1097/jcma.0000000000000400] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Due to the significance of preeclampsia (PE) and its adverse outcomes in the health of both mother and newborn, the present study was carried out to investigate the effect of aspirin on preventing the occurrence of PE, intrauterine growth restriction (IUGR), and preterm delivery in women with a previous history of PE. METHODS The present clinical trial was conducted on 90 pregnant women with a previous history of PE referred to the Khalij Fars Hospital in Bandar Abbas, Hormozgan Province Iran from April 2017 to August 2018. The subjects of the study were randomly assigned into two groups of intervention and control to receive either 80 mg of aspirin or placebo daily during the pregnancy. Patients' information was obtained and recorded upon entering the study, follow-up visits, and childbirth. RESULTS Among participants who entered the clinical trial, 86 patients (95.6%) completed the study. During the pregnancy, systolic blood pressure increased by 8.25 ± 14.83 and 19.06 ± 18.33 mmHg in aspirin and placebo groups, respectively (p = 0.001). Also, the same happened with diastolic blood pressure (6.12 ± 11.46 vs 13.48 ± 13.95 mmHg, p = 0.010). The rate of PE was equal to 27 (62.8%) and 38 (88.4%) in the aspirin and placebo groups, respectively (aOR = 0.23, p = 0.013). In the aspirin group, the rate of IUGR was equal to 27.9% compared with 25.6% of newborns in the control group (aOR = 1.18, p = 0.750). Similarly, there was no significant difference in the rate of preterm delivery between the two groups (p = 0.061). CONCLUSION The findings of the present study conducted exclusively on women with previous documented PE revealed that taking aspirin may have a preventive effect on PE in the current pregnancy.
Collapse
Affiliation(s)
- Nazanin Abdi
- Maternal-Fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Fertility and Infertility Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Afsane Rozrokh
- Student Research Committee, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Azin Alavi
- Fertility and Infertility Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Shahram Zare
- Epidemiology Department, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Homeira Vafaei
- Maternal-Fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasrin Asadi
- Maternal-Fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Kasraeian
- Maternal-Fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kamran Hessami
- Maternal-Fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Address correspondence. Dr. Kamran Hessami, Maternal-Fetal Medicine (Perinatology) Research Center, Shiraz University of Medical Sciences, Hafez Hospital, Chamran Ave., Shiraz, Iran. E-mail address: (K. Hessami)
| |
Collapse
|
41
|
Effect of High-Dose Intravenous Vitamin C on Postpartum Oxidative Stress in Severe Preeclampsia. REPRODUCTIVE MEDICINE 2020. [DOI: 10.3390/reprodmed1020009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Purpose: To determine whether high-dose intravenous vitamin C reduces oxidative stress in patients with severe preeclampsia in the first days postpartum. Methods: Biomarkers of oxidative stress were assessed as secondary outcomes of a single-center, randomized, placebo-controlled trial. Thirty-four patients with singleton pregnancies complicated by severe features of preeclampsia were randomized into two groups: intravenous vitamin C (1.5 g/6 h) (n = 17) or placebo (n = 17). Urinary concentrations of dityrosine, 8-hydroxy-2-deoxyguanosine (8-OHdg), 8-isoprostane, and N epsilon-(hexanoyl) lysine (HEL) were measured at days one and three after delivery and normalized for urinary creatinine in 22 of patients included (12 in vitamin C and 10 in placebo group). The Mann–Whitney U-test was used to compare values of oxidative stress biomarkers at days one and three after delivery in vitamin C vs. placebo groups (p ≤ 0.05 significant). Results: Dityrosine and 8-OHdg values did not differ significantly between the two study groups at day one after delivery (p = 0.23 and p = 0.77, respectively), but were significantly lower in the vitamin C group compared to the placebo group at day three after delivery (p = 0.04 and p = 0.03, respectively). Values of 8-isoprostane and HEL did not differ significantly between the two study groups at day one (p = 0.41 and p = 0.42, respectively), as well as at day three, after delivery (p = 0.25 and p = 0.24, respectively). Conclusion: High-dose intravenous vitamin C treatments in patients with severe preeclampsia reduced urinary levels of dityrosine and 8-OHdg (markers of protein and DNA oxidative damage, respectively) on day three after delivery. Vitamin C treatment had no significant effect on lipid peroxidation biomarkers, i.e., 8-isoprostane and HEL.
Collapse
|
42
|
Lorzadeh N, Kazemirad Y, Kazemirad N. Investigating the preventive effect of vitamins C and E on preeclampsia in nulliparous pregnant women. J Perinat Med 2020; 48:625-629. [PMID: 32755100 DOI: 10.1515/jpm-2019-0469] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/31/2020] [Indexed: 02/07/2023]
Abstract
Objectives Preeclampsia is a hypertensive disorder associated with pregnancy and is the leading cause of both maternal and neonatal morbidity and mortality. Recent reports suggest that free radical-induced endothelial cell injury might be an etiologic factor in the pathogenesis of preeclampsia. The aim of this study was to determine the protective effects of vitamins C and E for the prevention of preeclampsia. Methods This clinical trial was conducted in the year 2018 in the clinical centers of Lorestan University of Medical Sciences. One hundred and sixty nulliparous women aged 18-38 years without any risk factors for preeclampsia were divided into two groups. The first group received a 400-IU/day dose of vitamin E orally and a 1000-mg/day dose of vitamin C with iron tablets during 20-24 weeks of pregnancy, while the control group received only iron tablets. Finally, the data were analyzed using descriptive statistics and frequency distribution tables. Results The incidence of preeclampsia in the control group was 17.5% and in the intervention group was 5%, which was significantly different. The mean systolic pressures before and after intervention were 99.43 ± 7.8 and 12.44 ± 19.1, respectively, in the control group, and 99.3 ± 8.3 and 106.12 ± 13.25, respectively, in the intervention group (P < 0.001). The mean diastolic pressures before and after intervention in the control group were 62.7 ± 13.6 and 62.7 ± 4.7 (P < 0.001), respectively. Conclusion The results of this study show that vitamins C and E have protective effects against preeclampsia by alleviating the overall blood pressure.
Collapse
Affiliation(s)
- Nahid Lorzadeh
- Gynecology Department, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | | | - Nastran Kazemirad
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
Oxidative stress: Normal pregnancy versus preeclampsia. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165354. [DOI: 10.1016/j.bbadis.2018.12.005] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/22/2018] [Accepted: 12/05/2018] [Indexed: 02/03/2023]
|
44
|
The Pharmacokinetics of Vitamin C. Nutrients 2019; 11:nu11102412. [PMID: 31601028 PMCID: PMC6835439 DOI: 10.3390/nu11102412] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/08/2019] [Accepted: 10/08/2019] [Indexed: 02/07/2023] Open
Abstract
The pharmacokinetics of vitamin C (vitC) is indeed complex. Regulated primarily by a family of saturable sodium dependent vitC transporters (SVCTs), the absorption and elimination are highly dose-dependent. Moreover, the tissue specific expression levels and subtypes of these SVCTs result in a compartmentalized distribution pattern with a diverse range of organ concentrations of vitC at homeostasis ranging from about 0.2 mM in the muscle and heart, and up to 10 mM in the brain and adrenal gland. The homeostasis of vitC is influenced by several factors, including genetic polymorphisms and environmental and lifestyle factors such as smoking and diet, as well as diseases. Going from physiological to pharmacological doses, vitC pharmacokinetics change from zero to first order, rendering the precise calculation of dosing regimens in, for example, cancer and sepsis treatment possible. Unfortunately, the complex pharmacokinetics of vitC has often been overlooked in the design of intervention studies, giving rise to misinterpretations and erroneous conclusions. The present review outlines the diverse aspects of vitC pharmacokinetics and examines how they affect vitC homeostasis under a variety of conditions.
Collapse
|
45
|
Preeclampsia: Risk Factors, Diagnosis, Management, and the Cardiovascular Impact on the Offspring. J Clin Med 2019; 8:jcm8101625. [PMID: 31590294 PMCID: PMC6832549 DOI: 10.3390/jcm8101625] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 09/22/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
Hypertensive disorders of pregnancy affect up to 10% of pregnancies worldwide, which includes the 3%–5% of all pregnancies complicated by preeclampsia. Preeclampsia is defined as new onset hypertension after 20 weeks’ gestation with evidence of maternal organ or uteroplacental dysfunction or proteinuria. Despite its prevalence, the risk factors that have been identified lack accuracy in predicting its onset and preventative therapies only moderately reduce a woman’s risk of preeclampsia. Preeclampsia is a major cause of maternal morbidity and is associated with adverse foetal outcomes including intra-uterine growth restriction, preterm birth, placental abruption, foetal distress, and foetal death in utero. At present, national guidelines for foetal surveillance in preeclamptic pregnancies are inconsistent, due to a lack of evidence detailing the most appropriate assessment modalities as well as the timing and frequency at which assessments should be conducted. Current management of the foetus in preeclampsia involves timely delivery and prevention of adverse effects of prematurity with antenatal corticosteroids and/or magnesium sulphate depending on gestation. Alongside the risks to the foetus during pregnancy, there is also growing evidence that preeclampsia has long-term adverse effects on the offspring. In particular, preeclampsia has been associated with cardiovascular sequelae in the offspring including hypertension and altered vascular function.
Collapse
|
46
|
Liu T, Zhang M, Guallar E, Wang G, Hong X, Wang X, Mueller NT. Trace Minerals, Heavy Metals, and Preeclampsia: Findings from the Boston Birth Cohort. J Am Heart Assoc 2019; 8:e012436. [PMID: 31426704 PMCID: PMC6759885 DOI: 10.1161/jaha.119.012436] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022]
Abstract
Background Preeclampsia is a leading contributor to maternal and perinatal morbidity and mortality. In mice experiments, manganese (Mn) and selenium (Se) are protective whereas cadmium (Cd) is promotive for preeclampsia. Epidemiologic findings on these chemical elements have been inconsistent. To confirm experimental findings in mice, we examined associations of trace minerals (Mn and Se) and heavy metals (Cd, lead [Pb], and mercury [Hg]) with preeclampsia in a birth cohort. Methods and Results A total of 1274 women from the Boston Birth Cohort (enrolled since 1998) had complete data on the exposures and outcome. We measured Mn, Se, Cd, Pb, and Hg from red blood cells collected within 24 to 72 hours after delivery. We ascertained preeclampsia diagnosis from medical records. We used Poisson regression with robust variance models to estimate prevalence ratios (PRs) and 95% CIs. A total of 115 (9.0%) women developed preeclampsia. We observed evidence of a dose-response trend for Mn (P for trend<0.001) and to some extent for Cd (P for trend=0.009) quintiles. After multivariable adjustment, a 1 SD increment in Mn was associated with 32% lower risk of developing preeclampsia (PR=0.68; 95% CI, 0.54-0.86), whereas a 1 SD increment in Cd was associated with 15% higher risk of preeclampsia (PR=1.15; 95% CI, 0.98-1.36). Null associations were observed for Se, Pb, and Hg. Conclusions Findings from our cohort, consistent with evidence from mice experiments and human studies, indicate that women with lower blood concentration of Mn or higher Cd are more likely to develop preeclampsia.
Collapse
Affiliation(s)
- Tiange Liu
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
- Welch Center for Prevention, Epidemiology and Clinical ResearchJohns Hopkins UniversityBaltimoreMD
| | - Mingyu Zhang
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
| | - Eliseo Guallar
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
- Welch Center for Prevention, Epidemiology and Clinical ResearchJohns Hopkins UniversityBaltimoreMD
| | - Guoying Wang
- Center on the Early Life Origins of DiseaseDepartment of Population, Family and Reproductive HealthJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
| | - Xiumei Hong
- Center on the Early Life Origins of DiseaseDepartment of Population, Family and Reproductive HealthJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
| | - Xiaobin Wang
- Center on the Early Life Origins of DiseaseDepartment of Population, Family and Reproductive HealthJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
- Division of General Pediatrics & Adolescent MedicineDepartment of PediatricsJohns Hopkins School of MedicineBaltimoreMD
| | - Noel T. Mueller
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
- Welch Center for Prevention, Epidemiology and Clinical ResearchJohns Hopkins UniversityBaltimoreMD
| |
Collapse
|
47
|
Doğanlar ZB, Güçlü H, Öztopuz Ö, Türkön H, Dogan A, Uzun M, Doğanlar O. The Role of Melatonin in Oxidative Stress, DNA Damage, Apoptosis and Angiogenesis in Fetal Eye under Preeclampsia and Melatonin Deficiency Stress. Curr Eye Res 2019; 44:1157-1169. [PMID: 31090463 DOI: 10.1080/02713683.2019.1619778] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Aim: The aim of this study was to investigate the possible mechanisms of ocular damage induced by pinealectomy (PNX) and preeclampsia (PE), and to determine the cellular and molecular effects of melatonin treatment on oxidative stress, DNA damage, molecular chaperone responses, induction of apoptosis and angiogenesis in the fetal eye of both PNX and PNX+PE animals. Material and Methods: We analysed therapeutic potential of melatonin on fetal eye damage in PNX and PNX+PE animals using Malondialdehyde (MDA), Random Amplified Polymorphic DNA (RAPD), qRT-PCR and Western blot assays. Results: Our study presents three preliminary findings: (a) in fetal eye tissues, PNX and PNX+PE significantly induce oxidative damage to both DNA and protein contents, leading to a dramatic increase in caspase-dependent apoptotic signalling in both mitochondrial and death receptor pathways; (b) the same conditions trigger hypoxia biomarkers in addition to significant overexpression of HIF1-α, HIF1-β, MMP9 and VEGF genes in the fetal eye; (c) finally, melatonin regulates not only the expression of genes encoding antioxidant enzymes and increase in DNA damage as well as lipid peroxidation but also limits programmed cell death processes in the fetal eye of PNX and PNX+PE animals . Furthermore, melatonin can relatively modulate genes in the HIF1 family, TNF-α and VEGF, thus acting as a direct anti-angiogenic molecule. In conclusion, both PNX and PNX+PE induce ocular damage at both cellular and molecular levels in fetal eye tissue of rats. Conclusion: Our results clearly indicate the potential of melatonin as a preventative therapeutic intervention for fetal ocular damage triggered by both PNX and PNX+PE.
Collapse
Affiliation(s)
- Zeynep Banu Doğanlar
- Department of Medical Biology, Faculty of Medicine, Trakya University , Edirne , Turkey
| | - Hande Güçlü
- Department of Ophthalmology, Faculty of Medicine, Trakya University , Edirne , Turkey
| | - Özlem Öztopuz
- Department of Biophysics, Faculty of Medicine, Çanakkale Onsekiz Mart University , Çanakkale , Turkey
| | - Hakan Türkön
- Department of Biochemistry, Faculty of Medicine, Çanakkale Onsekiz Mart University , Çanakkale , Turkey
| | - Ayten Dogan
- Department of Medical Biology, Faculty of Medicine, Trakya University , Edirne , Turkey
| | - Metehan Uzun
- Department of Physiology, Faculty of Medicine, Çanakkale Onsekiz Mart University , Çanakkale , Turkey
| | - Oguzhan Doğanlar
- Department of Medical Biology, Faculty of Medicine, Trakya University , Edirne , Turkey
| |
Collapse
|
48
|
Cox AG, Gurusinghe S, Abd Rahman R, Leaw B, Chan ST, Mockler JC, Murthi P, Marshall SA, Lim R, Wallace EM. Sulforaphane improves endothelial function and reduces placental oxidative stress in vitro. Pregnancy Hypertens 2019; 16:1-10. [PMID: 31056142 DOI: 10.1016/j.preghy.2019.02.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/21/2019] [Accepted: 02/08/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The maternal endothelial dysfunction characteristic of preeclampsia arises, in part, from excessive placental production of anti-angiogenic factors, including soluble Flt-1, soluble endoglin and activin A, inducing oxidative stress. We assessed whether the antioxidant and NRF2-activator sulforaphane could mitigate endothelial and trophoblast dysfunction in vitro. METHODS We induced dysfunction in human umbilical vein endothelial cells (HUVECs) with TNF-α, assessing endothelial activation and dysfunction (endothelin-1, vascular cell adhesion molecule; VCAM1, intracellular adhesion molecule; ICAM1, e-selectin and endothelial permeability) in the presence or absence of sulforaphane. We also assessed the effects of sulforaphane in mitigating hypoxic and hyperoxic injury in term placental explants by measuring secretion of anti-angiogenic factors. To assess the role of NRF2 we silenced NRF2 in HUVECs and primary trophoblast cells. RESULTS Sulforaphane reduced TNF-α mediated HUVEC secretion of endothelin-1, VCAM1, ICAM1 and E-selectin, and prevented increased endothelial permeability. In placental explants, sulforaphane reduced the secretion of soluble Flt-1, soluble endoglin and activin A. Sulforaphane induced activation and nuclear translocation of NRF2 in HUVECs, inducing heme oxygenase 1. NRF2 silencing blocked some but not all of sulforaphane's effects in HUVECs. NRF2 silencing did not prevent sulforaphane's inhibition of trophobast secretion of soluble Flt-1 or activin A. CONCLUSION In reducing placental and endothelial oxidative stress, sulforaphane may offer a new adjuvant therapeutic approach for the treatment of preeclampsia.
Collapse
Affiliation(s)
- Annie G Cox
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia.
| | - Seshini Gurusinghe
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Rahana Abd Rahman
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia.
| | - Bryan Leaw
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
| | - Siow T Chan
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
| | - Joanne C Mockler
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia.
| | - Padma Murthi
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia.
| | - Sarah A Marshall
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia.
| | - Rebecca Lim
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
| | - Euan M Wallace
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
| |
Collapse
|
49
|
Cox AG, Marshall SA, Palmer KR, Wallace EM. Current and emerging pharmacotherapy for emergency management of preeclampsia. Expert Opin Pharmacother 2019; 20:701-712. [PMID: 30707633 DOI: 10.1080/14656566.2019.1570134] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Preeclampsia is a disease specific to pregnancy characterised by new onset hypertension with maternal organ dysfunction and/or fetal growth restriction. It remains a major cause of maternal and perinatal morbidity and mortality. For fifty years, antihypertensives have been the mainstay of treating preeclampsia, reducing maternal morbidity and mortality. With increased knowledge of the mechanisms underlying the disease has come opportunities for novel therapies that complement antihypertensives by protecting the maternal vasculature. Areas covered: In this review, the authors consider, in detail, the antihypertensives commonly used today in the emergency care of women with severe preeclampsia. They also review less common anti-hypertensive agents and discuss the role of magnesium sulphate in the management of preeclampsia and the prevention of eclampsia. Finally, they explore novel therapeutics for the acute management of preeclampsia. Expert opinion: The rapid control of maternal hypertension will, and must, remain the mainstay of emergency treatment for women with severe preeclampsia. The role of magnesium sulphate as a primary prevention for eclampsia is context dependant and should not displace a focus on correcting blood pressure safely. The exploration of novel adjuvant therapies will likely allow us to prolong pregnancy longer and improve perinatal outcomes safely for the mother.
Collapse
Affiliation(s)
- Annie G Cox
- a Ritchie Centre, Department of Obstetrics and GynaecologySchool of Clinical Sciences , Monash University , Clayton , Australia.,b The Ritchie Centre , Hudson Institute of Medical Research , Clayton , Australia
| | - Sarah A Marshall
- a Ritchie Centre, Department of Obstetrics and GynaecologySchool of Clinical Sciences , Monash University , Clayton , Australia.,b The Ritchie Centre , Hudson Institute of Medical Research , Clayton , Australia
| | - Kirsten R Palmer
- a Ritchie Centre, Department of Obstetrics and GynaecologySchool of Clinical Sciences , Monash University , Clayton , Australia
| | - Euan M Wallace
- a Ritchie Centre, Department of Obstetrics and GynaecologySchool of Clinical Sciences , Monash University , Clayton , Australia.,b The Ritchie Centre , Hudson Institute of Medical Research , Clayton , Australia
| |
Collapse
|
50
|
WANG Z, WANG C, QIU J, NI Y, CHAI S, ZHOU L, LI J, YAN B, YANG J, LIU Q. The Association between Dietary Vitamin C/E and Gestational Hypertensive Disorder: A Case-Control Study. J Nutr Sci Vitaminol (Tokyo) 2018; 64:454-465. [DOI: 10.3177/jnsv.64.454] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
| | - Cheng WANG
- Gansu Provincial Maternity and Child-care Hospital
| | - Jie QIU
- Gansu Provincial Maternity and Child-care Hospital
| | - Yali NI
- Gansu Provincial Maternity and Child-care Hospital
| | - Sanming CHAI
- Gansu Provincial Maternity and Child-care Hospital
| | - Li ZHOU
- Gansu Provincial Maternity and Child-care Hospital
| | - Jing LI
- Gansu Provincial Maternity and Child-care Hospital
| | - Bo YAN
- Gansu Provincial Maternity and Child-care Hospital
| | - Jie YANG
- Gansu Provincial Maternity and Child-care Hospital
| | - Qing LIU
- Department of Gynecology and Obstetrics of Gansu Provincial Maternity and Child-care Hospital
| |
Collapse
|