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
|
Nguyen P, Sanderson B, Makama M, Mills K, Ammerdorffer A, Gülmezoglu AM, Vogel JP, McDougall ARA. Polyphenols for the Prevention or Management of Preeclampsia: A Systematic Review and Meta-Analysis. BJOG 2025; 132:867-879. [PMID: 40025969 PMCID: PMC12051244 DOI: 10.1111/1471-0528.18106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 03/04/2025]
Abstract
OBJECTIVES To evaluate the effects of polyphenol-containing products during pregnancy on preeclampsia-related maternal and neonatal outcomes. DESIGN Systematic review and meta-analysis. SETTING Nine databases and one trial registry, from inception to August 11th, 2023. POPULATION/SAMPLE Randomised controlled trials where women received polyphenolic-containing products (as standardised extracts or dietary supplements) compared to placebo or standard care. METHODS All review stages were conducted by two independent reviewers. Random-effects meta-analysis with the Hartung-Knapp-Sidik-Jonkman method using a framework for studies with few events. MAIN OUTCOME MEASURES Clinical outcomes combining the core outcome set for preeclampsia and WHO's priority outcomes. RESULTS Fourteen trials investigating six candidates were included. In women with preeclampsia, the addition of epigallocatechin gallate (EGCG) to nifedipine may reduce the time needed to achieve blood pressure control (mean difference (MD) = -14.10 min, 95% CI -18.46 to -9.74) and increase the time to the next hypertensive crisis (MD = 3.10 h, 95% CI 2.35 to 3.85) compared to nifedipine alone (1 trial, 349 women; low certainty). Similarly, the addition of resveratrol to nifedipine may reduce the time needed to achieve blood pressure control (MD = -15.50 min, 95% CI -19.83 to -11.17) and increase the time to the next hypertensive crisis (MD = 2.50 h, 95% CI 2.09 to 2.91) (1 trial, 349 women; low certainty). No differences were observed for other outcomes or candidates (Salvia miltiorrhiza, Bryophyllum pinnatum , raspberry and cranberry extracts). CONCLUSIONS ECGC and resveratrol supplements have been investigated for potential effects in managing clinical signs and symptoms of preeclampsia; however, evidence on the clinical and adverse effects of polyphenols is limited and uncertain.
Collapse
Affiliation(s)
- Phi‐Yen Nguyen
- Maternal, Child and Adolescent Health ProgramBurnet InstituteMelbourneAustralia
| | - Ben Sanderson
- Maternal, Child and Adolescent Health ProgramBurnet InstituteMelbourneAustralia
| | - Maureen Makama
- Maternal, Child and Adolescent Health ProgramBurnet InstituteMelbourneAustralia
| | - Kate Mills
- Maternal, Child and Adolescent Health ProgramBurnet InstituteMelbourneAustralia
| | | | | | - Joshua P. Vogel
- Maternal, Child and Adolescent Health ProgramBurnet InstituteMelbourneAustralia
| | | |
Collapse
|
3
|
Gladstone RA, Snelgrove JW, McLaughlin K, Hobson SR, Windrim RC, Melamed N, Hladunewich M, Drewlo S, Kingdom JC. Placental growth factor (PlGF) and soluble fms-like tyrosine kinase-1 (sFlt1): powerful new tools to guide obstetric and medical care in pregnancy. Obstet Med 2025:1753495X251327462. [PMID: 40191640 PMCID: PMC11969481 DOI: 10.1177/1753495x251327462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 01/02/2025] [Accepted: 01/14/2025] [Indexed: 04/09/2025] Open
Affiliation(s)
- Rachel A Gladstone
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynaecology, Mount Sinai Hospital, Canada
| | - John W Snelgrove
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynaecology, Mount Sinai Hospital, Canada
| | - Kelsey McLaughlin
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynaecology, Mount Sinai Hospital, Canada
| | - Sebastian R Hobson
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynaecology, Mount Sinai Hospital, Canada
| | - Rory C Windrim
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynaecology, Mount Sinai Hospital, Canada
| | - Nir Melamed
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynaecology, Sunnybrook Health Sciences Centre, University of Toronto, Canada
| | - Michelle Hladunewich
- Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Canada
| | - Sascha Drewlo
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Canada
| | - John C Kingdom
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynaecology, Mount Sinai Hospital, Canada
| |
Collapse
|
4
|
Li S, Zhu J, Zhao Y, An P, Zhao H, Xiong Y. Metabolic disorder of nutrients-an emerging field in the pathogenesis of preeclampsia. Front Nutr 2025; 12:1560610. [PMID: 40123939 PMCID: PMC11925777 DOI: 10.3389/fnut.2025.1560610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/24/2025] [Indexed: 03/25/2025] Open
Abstract
It is well acknowledged that metabolic disorder binds closely with preeclampsia, though some of the causal relationships are still ambiguous. This review systematically summarizes the metabolic characteristics of carbohydrates, lipids, amino acids, and glycans in preeclampsia, highlighting their roles in oxidative stress, trophoblast autophagy, inflammatory response, and vascular tone regulation. Key findings include upregulated glycolysis and impaired mitochondrial function contributing to ATP deficiency, dysregulated lipid metabolism exacerbating oxidative stress and vascular dysfunction, and amino acid imbalances disrupting immune responses and redox homeostasis. Emerging therapies, such as metformin and pravastatin, demonstrate potential in targeting these pathways for prevention and treatment. Here, we reviewed thoroughly the related literature with a view to delineating the potential association of nutrient metabolism with preeclampsia, so that we could explore a promising therapeutic approach.
Collapse
Affiliation(s)
- Shuyue Li
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- The Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Jie Zhu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- The Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Ying Zhao
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- The Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Ping An
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- The Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Huanqiang Zhao
- Shenzhen Maternal and Child Health Hospital, Shenzhen, China
| | - Yu Xiong
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- The Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| |
Collapse
|
5
|
Anness AR, Khalil A, Mousa HA. Arterial stiffness in gestational diabetes: Latest insights. Best Pract Res Clin Obstet Gynaecol 2025; 99:102587. [PMID: 39970706 DOI: 10.1016/j.bpobgyn.2025.102587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 01/29/2025] [Indexed: 02/21/2025]
Abstract
Increased arterial stiffness is a known cardiovascular risk factor, associated with hypertension and acute coronary events. Gestational diabetes (GDM) is associated with the development of placental-mediated disorders and future cardiovascular morbidity, raising the possibility of an association with increased arterial stiffness (AS). Several studies have now investigated this association through the assessment of pulse wave velocity and augmentation index. In the current review, we present the latest evidence regarding the changes in arterial stiffness in pregnancies complicated by GDM, before the onset of clinical disease, during its course, and after its resolution. We also review the evidence that AS could influence the need for different treatments for GDM, and the impact that the treatments, and in particular, metformin, could have on arterial stiffness.
Collapse
Affiliation(s)
- Abigail R Anness
- Princess Anne Hospital, University Hospitals of Southampton, UK.
| | - Asma Khalil
- St. George's University Hospital (University of London), UK
| | - Hatem A Mousa
- Maternal and Fetal Medicine Unit, University Hospitals of Leicester NHS Trust, UK; University of Leicester, UK
| |
Collapse
|
6
|
May L, Mason D, van de Vyver M, Conradie M, Hall DR. Hypertensive disorders in a gestational diabetes cohort from Cape Town, South Africa. Pregnancy Hypertens 2025; 39:101185. [PMID: 39855048 DOI: 10.1016/j.preghy.2025.101185] [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: 10/16/2024] [Revised: 12/19/2024] [Accepted: 01/05/2025] [Indexed: 01/27/2025]
Abstract
INTRODUCTION AND OBJECTIVE As thresholds for the diagnosis of gestational diabetes mellitus (GDM) become lower, and the prevalence of obesity in society rises, more pregnant women will be diagnosed with GDM and hypertension. Both conditions hold dangers for mother and baby. Our objective was to properly describe this association. STUDY DESIGN AND OUTCOME MEASURES This retrospective audit was conducted at the Obstetric High-Risk Clinic at Tygerberg Academic Hospital, Cape Town, South Africa. All patients diagnosed with GDM from 01/01/19 - 31/12/19 were included. The primary aim was to determine the incidence, and different classes of hypertensive disease associated with GDM in this population. The secondary aims were to evaluate associations with the development of hypertensive disorders in women with GDM and to include descriptions of the management, course and complications during the pregnancies, including the early neonatal period. The research was approved by the Ethics Committee of Stellenbosch University. RESULTS Of the 274 women with GDM, included in the analysis, 204 (75 %) had concomitant hypertension. Classes of hypertension were chronic hypertension 21 % (58/274), gestational hypertension 26 % (71/274), and pre-eclampsia (de novo and super-imposed) 27 % (75/275). Those without hypertension had significantly lower BMIs: 35 [29-42] vs 41 [34-45] kg/m2 (median [IQR]; p < 0.001). Using ROC curves, BMI at booking had strong associations with chronic and gestational hypertension (p = 0,002; p = 0,001), and pre-eclampsia (p = 0,002). All three intra-uterine deaths (two spontaneous and one iatrogenic) occurred in the GDM + hypertension group. CONCLUSION Hypertensive conditions of pregnancy were common amongst a referral-based population with GDM, with BMI being the strongest predictor.
Collapse
Affiliation(s)
- L May
- Department of Obstetrics & Gynaecology Stellenbosch University and Tygerberg Hospital Cape Town South Africa
| | - D Mason
- Department of Obstetrics & Gynaecology Stellenbosch University and Tygerberg Hospital Cape Town South Africa
| | - M van de Vyver
- Department of Medicine Experimental Medicine Research Group Stellenbosch University Cape Town South Africa
| | - M Conradie
- Department of Medicine Division of Endocrinology Stellenbosch University and Tygerberg Hospital Cape Town South Africa
| | - D R Hall
- Department of Obstetrics & Gynaecology Stellenbosch University and Tygerberg Hospital Cape Town South Africa.
| |
Collapse
|
7
|
Afrose D, Johansen MD, Nikolic V, Karadzov Orlic N, Mikovic Z, Stefanovic M, Cakic Z, Hansbro PM, McClements L. Evaluating oxidative stress targeting treatments in in vitro models of placental stress relevant to preeclampsia. Front Cell Dev Biol 2025; 13:1539496. [PMID: 40109359 PMCID: PMC11920713 DOI: 10.3389/fcell.2025.1539496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 01/20/2025] [Indexed: 03/22/2025] Open
Abstract
Background Preeclampsia is a complex pregnancy disorder characterized by the new onset of hypertension and organ dysfunction, often leading to significant maternal and fetal morbidity and mortality. Placental dysfunction is a hallmark feature of preeclampsia, which is often caused by inappropriate trophoblast cell function in association with oxidative stress, inflammation and/or pathological hypoxia. This study explores the role of oxidative stress in trophoblast cell-based models mimicking the preeclamptic placenta and evaluates potential therapeutic strategies targeting these mechanisms. Methods Uric acid (UA) and malondialdehyde (MDA) concentrations were measured in human plasma from women with preeclampsia (n = 24) or normotensive controls (n = 14) using colorimetric assays. Custom-made first trimester trophoblast cell line, ACH-3P, was exposed to various preeclampsia-like stimuli including hypoxia mimetic (dimethyloxalylglycine or DMOG, 1 mM), inflammation (tumour necrosis factor or TNF-α, 10 ng/mL) or mitochondria dysfunction agent, (Rhodamine-6G or Rho-6G, 1 μg/mL), ± aspirin (0.5 mM), metformin (0.5 mM), AD-01 (100 nM) or resveratrol (15 µM), for 48 h. Following treatments, UA/MDA, proliferation (MTT), wound scratch and cytometric bead, assays, were performed. Results Overall, MDA plasma concentration was increased in the preeclampsia group compared to healthy controls (p < 0.001) whereas UA showed a trend towards an increase (p = 0.06); when adjusted for differences in gestational age at blood sampling, MDA remained (p < 0.001) whereas UA became (p = 0.03) significantly correlated with preeclampsia. Our 2D first trimester trophoblast cell-based in vitro model of placental stress as observed in preeclampsia, mimicked the increase in UA concentration following treatment with DMOG (p < 0.0001), TNF-α (p < 0.05) or Rho-6G (p < 0.001) whereas MDA cell concentration increased only in the presence of DMOG (p < 0.0001) or Rho-6G (p < 0.001). Metformin was able to abrogate DMOG- (p < 0.01), Rho-6G- (p < 0.0001) or TNF-α- (p < 0.01) induced increase in UA, or DMOG- (p < 0.0001) or TNF-α- (p < 0.05)induced increase in MDA. AD-01 abrogated UA or MDA increase in the presence of TNF-α (p < 0.001) or Rho-6G (p < 0.001)/DMOG (p < 0.0001), respectively. The preeclampsia-like stimuli also mimicked adverse impact on trophoblast cell proliferation, migration and inflammation, most of which were restored with either aspirin, metformin, resveratrol, or AD-01 (p < 0.05). Conclusion Our 2D in vitro models recapitulate the response of the first trimester trophoblast cells to preeclampsia-like stresses, modelling inappropriate placental development, and demonstrate therapeutic potential of repurposed treatments.
Collapse
Affiliation(s)
- Dinara Afrose
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Matt D Johansen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Valentina Nikolic
- Department of Pharmacology with Toxicology, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Natasa Karadzov Orlic
- Department of Gynaecology and Obstetrics, Narodni Front, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Zeljko Mikovic
- Department of Gynaecology and Obstetrics, Narodni Front, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milan Stefanovic
- Department of Gynaecology and Obstetrics, Clinical Centre Nis, Nis, Serbia
- Department of Gynaecology and Obstetrics, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Zoran Cakic
- Department of Gynaecology and Obstetrics, General Hospital of Leskovac, Leskovac, Serbia
| | - Philip M Hansbro
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- Centre for Inflammation, Centenary Institute and 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
|
8
|
Onda K. [Translational Research on Pre-eclampsia with Existing Drugs Targeting Antioxidant Molecules]. YAKUGAKU ZASSHI 2025; 145:43-47. [PMID: 39756924 DOI: 10.1248/yakushi.24-00174-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Pre-eclampsia, a type of hypertensive disorders of pregnancy (HDP), is characterized by hypertension and organ dysfunction that develops or worsens after 20 weeks of gestation. Although symptomatic management using antihypertensive medications has been adopted, definitive treatments other than pregnancy termination remain unavailable to halt disease progression. Research on heme oxygenase (HO)-1, a molecule with anti-inflammatory and antioxidative properties, has shown that a pharmacological increase in placental HO-1 expression and activity may ameliorate this condition; therefore, HO-1 is a promising therapeutic target for this disorder. Medications with properties that can be used during pregnancy are strong candidates for repurposing. In this article, I discuss the potential applications of proton pump inhibitors in the prevention or treatment of preeclampsia by presenting our foundational research and subsequent observational and interventional clinical studies.
Collapse
Affiliation(s)
- Kenji Onda
- Department of Clinical Pharmacology, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| |
Collapse
|
9
|
Heath-Freudenthal A, Estrada A, von Alvensleben I, Julian CG. Surviving birth at high altitude. J Physiol 2024; 602:5463-5473. [PMID: 38520695 PMCID: PMC11418585 DOI: 10.1113/jp284554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 03/11/2024] [Indexed: 03/25/2024] Open
Abstract
This Symposium Review examines challenges to surviving birth and infancy at high altitudes. Chronic exposure to the environmental hypoxia of high altitudes increases the incidence of maternal vascular disorders of pregnancy characterized by placental insufficiency, restricted fetal growth and preterm delivery, and impairs pulmonary vascular health during infancy. While each condition independently contributes to excess morbidity and mortality in early life, evidence indicates vascular disorders of pregnancy and infantile pulmonary vascular dysfunction are intertwined. By integrating our recent scientific and clinical observations in Bolivia with existing literature, we propose potential avenues to reduce the infant mortality burden at high altitudes and reduce pulmonary vascular disease in highland neonates, and emphasize the need for further research to address unresolved questions.
Collapse
Affiliation(s)
| | | | | | - Colleen G. Julian
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, US
| |
Collapse
|
10
|
Patel M, Battarbee AN, Refuerzo JS, Zork N, Eichelberger K, Ramos GA, Olson G, Durnwald C, Landon MB, Aagaard KM, Wallace K, Scifres C, Rosen T, Mulla W, Valent A, Longo S, Boggess KA. Association Between Metformin Use in Early Gestational or Type 2 Diabetes in Pregnancy and Preterm Preeclampsia. Obstet Gynecol 2024; 144:733-739. [PMID: 39236318 PMCID: PMC11575948 DOI: 10.1097/aog.0000000000005720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 07/25/2024] [Indexed: 09/07/2024]
Abstract
OBJECTIVE To estimate the association between maternal metformin use for the treatment of early gestational or pre-existing type 2 diabetes and preterm preeclampsia. METHODS This is a planned secondary analysis of the MOMPOD study (Medical Optimization of Management of Overt Type 2 Diabetes in Pregnancy), a randomized trial comparing the effect of adding metformin with insulin treatment on composite neonatal outcome in singleton pregnancies with early gestational or type 2 diabetes. Participants were randomized at 11-23 weeks of gestation to 1,000 mg metformin twice daily or placebo until delivery. A subset of participants had maternal blood collected at 24-30 weeks of gestation, and serum soluble endoglin, apolipoprotein B, vascular cell adhesion molecule-1, soluble fms-like tyrosine kinase 1, placental growth factor, high-sensitivity C-reactive protein, adiponectin, and vascular endothelial growth factor levels were measured. Our primary outcome was preterm preeclampsia , defined as preeclampsia requiring delivery before 37 weeks of gestation. Secondary outcomes included preterm preeclampsia requiring delivery before 34 weeks of gestation and differences in serum biomarkers. Multivariable regression analysis was used to estimate the associations between metformin use and primary or secondary study outcomes. RESULTS Of 831 participants, 119 (14.3%) developed preeclampsia requiring delivery before 37 weeks of gestation: 57 of 416 (13.7%) in the placebo group and 62 of 415 (14.9%) in the metformin group. Thirty-seven (4.4%) developed preeclampsia requiring delivery before 34 weeks of gestation: 15 (3.6%) receiving placebo and 22 (5.3%) receiving metformin. Compared with placebo, metformin was not associated with a significant difference in the occurrence of preeclampsia before 37 weeks of gestation (adjusted odds ratio [aOR] 1.04, 95% CI, 0.70-1.56) or before 34 weeks (aOR 1.43, 95% CI, 0.73-2.81). Similarly, there was no association between maternal metformin use and serum biomarker levels. CONCLUSION Among parturients with early gestational or pre-existing type 2 diabetes, the addition of metformin to insulin was not associated with lower odds of preterm preeclampsia or with serum biomarkers associated with cardiovascular disease risk.
Collapse
Affiliation(s)
- Maya Patel
- University of North Carolina at Chapel Hill School of Medicine Chapel Hill, NC USA
| | - Ashley N. Battarbee
- University of Alabama at Birmingham Heersink School of Medicine Birmingham, AL USA
| | - Jerrie S. Refuerzo
- University of Texas Health Houston McGovern Medical School Houston, TX USA
| | - Noelia Zork
- Columbia University Irving Medical Center, New York, NY USA
| | - Kacey Eichelberger
- University of South Carolina School of Medicine Greenville/Prisma Health-Upstate Greenville, SC USA
| | | | - Gayle Olson
- University of Texas Medical Branch Galveston Galveston, TX USA
| | - Celeste Durnwald
- University of Pennsylvania Perelman School of Medicine Philadelphia, PA USA
| | - Mark B. Landon
- Ohio State University College of Medicine and Wexner Medical Center Columbus, OH USA
| | - Kjersti M. Aagaard
- Baylor College of Medicine and Texas Children’s Hospital Houston, TX USA
| | - Kedra Wallace
- University of Mississippi Medical Center Jackson, MS USA
| | | | - Todd Rosen
- Rutgers Health/Robert Wood Johnson Medical School New Brunswick, NJ USA
| | - Wadia Mulla
- Temple University Lewis Katz School of Medicine Philadelphia, PA USA
| | - Amy Valent
- Oregon Health & Science University Portland, OR USA
| | | | - Kim A. Boggess
- University of North Carolina at Chapel Hill School of Medicine Chapel Hill, NC USA
| |
Collapse
|
11
|
Gordon H, Salim N, Tong S, Walker S, De Silva M, Cluver C, Mehdipour P, Hiscock R, Sutherland L, Doust A, Bergman L, Wikström AK, Lindquist A, Hesselman S, Hastie R. Metformin use and preeclampsia risk in women with diabetes: a two-country cohort analysis. BMC Med 2024; 22:418. [PMID: 39334302 PMCID: PMC11438264 DOI: 10.1186/s12916-024-03628-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Metformin is a hypoglycaemic medication that has been proposed to treat or prevent preeclampsia. Combining national birth data from Scotland and Sweden, we investigated whether metformin used during pregnancy was associated with an altered risk of developing a hypertensive disorder of pregnancy. METHODS We utilised data from two population-based cohorts: Scotland (2012-2018) and Sweden (2007-2019). Nulliparous women with gestational diabetes or type 2 diabetes who had birth outcome data linked with medications prescribed during pregnancy were included. The association between metformin prescription and hypertensive disorders of pregnancy was characterised using inverse probability weighted regression analysis, adjusting for variables that predict metformin use and potential confounders. Adverse neonatal outcomes were included as secondary outcomes. Results from both countries were then combined in a meta-analysis using a random effects model. RESULTS The Scottish cohort included 3859 women with gestational diabetes or type 2 diabetes. Of these women, 30.8% (n = 1187) received at least one metformin prescription during pregnancy. For Sweden, 7771 women with gestational diabetes were included where 19.3% (1498) used metformin during pregnancy. Metformin prescription was not associated with an altered risk of any hypertensive disorder of pregnancy (Scotland adjusted relative risk (aRR) 0.88 [95% confidence interval (CI) 0.66-1.19]; Sweden aRR 1.08 [95% CI 0.86-1.37]) or preeclampsia (Scotland aRR 1.02 [95% CI 0.66-1.60]; Sweden aRR 1.00 [95% CI 0.72-1.39]). Combining adjusted results in a meta-analysis produced similar findings, with a pooled RR of 0.98 (95% CI 0.79-1.18) for any hypertensive disorder and RR 1.01 ([95% CI 0.73-1.28]) for preeclampsia. For neonatal outcomes, metformin was associated with a reduced risk of birthweight > 4500 g in Scotland (aRR 0.39 [95% CI 0.21-0.71]) but not in Sweden. There was no association between metformin and preterm birth or birthweight < 3rd or < 10th percentiles. Pooling results from both countries, metformin was not associated with adverse neonatal outcomes, including preterm birth (RR 1.00 [95% CI 0.89-1.13]), and birthweight < 10th percentile (RR 0.82 [95% CI 0.60-1.13]) or < 3rd percentile (RR 0.78 [95% CI 0.41-1.48]). CONCLUSIONS In this two-country analysis, metformin use in pregnancy among women with diabetes was not associated with an altered risk of developing any hypertensive disorder of pregnancy. In the combined meta-analysis, metformin was not associated with an altered risk of adverse neonatal outcomes.
Collapse
Affiliation(s)
- Hannah Gordon
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Melbourne, Australia.
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC, Australia.
| | - Noor Salim
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Stephen Tong
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Melbourne, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC, Australia
| | - Susan Walker
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Melbourne, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC, Australia
| | - Manarangi De Silva
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Melbourne, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC, Australia
| | - Catherine Cluver
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Melbourne, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC, Australia
- Department of Obstetrics and Gynecology, Stellenbosch University, Cape Town, South Africa
| | - Parinaz Mehdipour
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Melbourne, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC, Australia
| | - Richard Hiscock
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Melbourne, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC, Australia
| | - Lauren Sutherland
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Ann Doust
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Lina Bergman
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
- Department of Obstetrics and Gynecology, Stellenbosch University, Cape Town, South Africa
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Anna-Karin Wikström
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Anthea Lindquist
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Melbourne, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC, Australia
| | - Susanne Hesselman
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
- Centre for Clinical Research, Uppsala University, Falun, Sweden
| | - Roxanne Hastie
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Melbourne, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC, Australia
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| |
Collapse
|
12
|
Elgazzaz M, Woodham PC, Maher J, Faulkner JL. Implications of pregnancy on cardiometabolic disease risk: preeclampsia and gestational diabetes. Am J Physiol Cell Physiol 2024; 327:C646-C660. [PMID: 39010840 PMCID: PMC11427017 DOI: 10.1152/ajpcell.00293.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024]
Abstract
Cardiometabolic disorders, such as obesity, insulin resistance, and hypertension, prior to and within pregnancy are increasing in prevalence worldwide. Pregnancy-associated cardiometabolic disease poses a great risk to the short- and long-term well-being of the mother and offspring. Hypertensive pregnancy, notably preeclampsia, as well as gestational diabetes are the major diseases of pregnancy growing in prevalence as a result of growing cardiometabolic disease prevalence. The mechanisms whereby obesity, diabetes, and other comorbidities lead to preeclampsia and gestational diabetes are incompletely understood and continually evolving in the literature. In addition, novel therapeutic avenues are currently being explored in these patients to offset cardiometabolic-induced adverse pregnancy outcomes in preeclamptic and gestational diabetes pregnancies. In this review, we discuss the emerging pathophysiological mechanisms of preeclampsia and gestational diabetes in the context of cardiometabolic risk as well as the most recent preclinical and clinical updates in the pathogenesis and treatment of these conditions.
Collapse
Affiliation(s)
- Mona Elgazzaz
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Padmashree C Woodham
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - James Maher
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Jessica L Faulkner
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
13
|
Cluver CA, Bergman L, Imberg H, Mol BW, Hall D, Bekker A, Gordon A, Brownfoot F, Kaitu'u-Lino TJ, Walker SP, Tong S. Does metformin prolong pregnancy in preterm pre-eclampsia? A study protocol for a South African, hospital-based double-blind, randomised, placebo-controlled trial. BMJ Open 2024; 14:e082880. [PMID: 38890136 PMCID: PMC11191812 DOI: 10.1136/bmjopen-2023-082880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
INTRODUCTION Preterm pre-eclampsia is a leading cause of maternal morbidity and mortality. The Pre-eclampsia Intervention 2 (PI 2) trial suggested that metformin sustained release (XR) may prolong gestation by a week in pregnant women undergoing expectant management (7.6 days, geometric mean ratio 1.39, 95% CI 0.99 to 1.95; p=0.057). These findings should be confirmed with a larger sample size, and we need to know if such a prolongation improves neonatal outcome. Here, we describe the protocol for such a follow-up trial. METHODS The PI 3 trial is a phase III, intention-to-treat, double-blind, placebo-controlled randomised clinical trial to assess if metformin XR can prolong gestation and improve neonatal outcomes in women undergoing expectant management for preterm pre-eclampsia. We will recruit women who are between 26+0 and 31+6 weeks pregnant. Women will be randomised to receive either 3 g metformin XR or an identical placebo in divided daily doses. The primary outcome is prolongation of pregnancy. Secondary outcomes are neonatal birth weight and length of neonatal care admission (an indicator of neonatal health at birth). All other outcomes will be exploratory. We will record tolerability and adverse events. We plan a sample size of 500 participants to be powered for the primary and secondary outcomes. ETHICS AND DISSEMINATION PI 3 has ethical approval (Health Research Ethics Committee 2, Stellenbosch University, Protocol number M21/03/007, Project ID 21639, Federal Wide Assurance Number 00001372, Institutional Review Board Number IRB0005239), and is registered with the Pan African Clinical Trial Registry (PACTR202104532026017) and the South African Medicine Control Council (20211211). Data will be presented at international conferences and published in peer-reviewed journals. TRIAL REGISTRATION NUMBER PACTR202104532026017).
Collapse
Affiliation(s)
- Catherine Anne Cluver
- Department of Obstetrics and Gynaecology, Stellenbosch University, Faculty of Medicine and Health Sciences, Cape Town, South Africa
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, Melbourne University, Heidelberg, Victoria, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia
| | - Lina Bergman
- Department of Obstetrics and Gynaecology, Stellenbosch University, Faculty of Medicine and Health Sciences, Cape Town, South Africa
- Department of Obstetrics and Gynaecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Ben W Mol
- Department of Obstetrics and Gynaecology, Monash Medical School, Monash University, Melbourne, Victoria, Australia
| | - David Hall
- Department of Obstetrics and Gynaecology, Stellenbosch University, Faculty of Medicine and Health Sciences, Cape Town, South Africa
| | - Adrie Bekker
- Department of Paediatrics, Stellenbosch University, Faculty of Medicine and Health Sciences, Cape Town, South Africa
| | - Adrienne Gordon
- Discipline of Obstetrics, Gynaecology and Neonatology, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Fiona Brownfoot
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, Melbourne University, Heidelberg, Victoria, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, Melbourne University, Heidelberg, Victoria, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia
| | - Susan P Walker
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, Melbourne University, Heidelberg, Victoria, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia
| | - Stephen Tong
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, Melbourne University, Heidelberg, Victoria, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia
- Translational Obstetrics Group and Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, Heidelberg, Melbourne, Victoria, Australia
| |
Collapse
|
14
|
Ngene NC, Moodley J. Preventing maternal morbidity and mortality from preeclampsia and eclampsia particularly in low- and middle-income countries. Best Pract Res Clin Obstet Gynaecol 2024; 94:102473. [PMID: 38513504 DOI: 10.1016/j.bpobgyn.2024.102473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/15/2023] [Accepted: 02/05/2024] [Indexed: 03/23/2024]
Abstract
Preeclampsia (PE) is a complex heterogeneous disorder with overlapping clinical phenotypes that complicate diagnosis and management. Although several pathophysiological mechanisms have been proposed, placental dysfunction due to inadequate remodelling of uterine spiral arteries leading to mal-perfusion and syncytiotrophoblast stress is recognized as the unifying characteristic of early-onset PE. Placental overgrowth and or premature senescence are probably the causes of late-onset PE. The frequency of PE has increased over the last few decades due to population-wide increases in risk factors viz. obesity, diabetes, multifetal pregnancies and pregnancies at an advanced maternal age. Whilst multimodal tools with components comprising risk factors, biomarkers and sonography are used for predicting PE, aspirin is most effective in preventing early-onset PE. The incidence and clinical consequences of PE and eclampsia are influenced by socioeconomic and cultural factors, therefore management strategies should involve multi-sector partnerships to mitigate the adverse outcomes.
Collapse
Affiliation(s)
- Nnabuike Chibuoke Ngene
- Department of Obstetrics and Gynaecology, Rahima Moosa Mother and Child Hospital, Johannesburg, Gauteng, South Africa; Department of Obstetrics and Gynaecology, School of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, Gauteng, South Africa.
| | - Jagidesa Moodley
- Women's Health and HIV Research Group, Department of Obstetrics and Gynecology, School of Clinical Medicine, Faculty of Health Sciences, University of Kwa Zulu-Natal, Durban, South Africa.
| |
Collapse
|
15
|
Brownfoot F, Rolnik DL. Prevention of preeclampsia. Best Pract Res Clin Obstet Gynaecol 2024; 93:102481. [PMID: 38373378 DOI: 10.1016/j.bpobgyn.2024.102481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/19/2023] [Accepted: 02/05/2024] [Indexed: 02/21/2024]
Abstract
Preeclampsia is a relatively common pregnancy complication and constitutes a major cause of morbidity and mortality for mothers and children worldwide. It disproportionally affects low-resource countries. Appropriate identification of individuals at increased risk and prevention of the disease and its complications remain healthcare and research priorities, and the investigation of potential interventions to prevent preeclampsia has driven much of the obstetric research in recent decades. In this article, we review the scientific literature on the topic, highlighting established benefits and remaining questions regarding different non-pharmacological and pharmacological strategies, including exercise, the timing of birth, aspirin and calcium use, among others, as well as potential novel therapies under investigation.
Collapse
Affiliation(s)
- Fiona Brownfoot
- Mercy Hospital for Women, Heidelberg, Victoria, Australia; Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia
| | - Daniel Lorber Rolnik
- Women's and Newborn, Monash Health, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
16
|
Thadhani R, Cerdeira AS, Karumanchi SA. Translation of mechanistic advances in preeclampsia to the clinic: Long and winding road. FASEB J 2024; 38:e23441. [PMID: 38300220 DOI: 10.1096/fj.202301808r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/30/2023] [Accepted: 01/09/2024] [Indexed: 02/02/2024]
Abstract
As one of the leading causes of premature birth and maternal and infant mortality worldwide, preeclampsia remains a major unmet public health challenge. Preeclampsia and related hypertensive disorders of pregnancy are estimated to cause >75 000 maternal and 500 000 infant deaths globally each year. Because of rising rates of risk factors such as obesity, in vitro fertilization and advanced maternal age, the incidence of preeclampsia is going up with rates ranging from 5% to 10% of all pregnancies worldwide. A major discovery in the field was the realization that the clinical phenotypes related to preeclampsia, such as hypertension, proteinuria, and other adverse maternal/fetal events, are due to excess circulating soluble fms-like tyrosine kinase-1 (sFlt-1, also referred to as sVEGFR-1). sFlt-1 is an endogenous anti-angiogenic protein that is made by the placenta and acts by neutralizing the pro-angiogenic proteins vascular endothelial growth factor (VEGF) and placental growth factor (PlGF). During the last decade, this work has spawned a new era of molecular diagnostics for early detection of this condition. Antagonizing sFlt-1 either by reducing production or blocking its actions has shown salutary effects in animal models. Further, in early-stage human studies, the therapeutic removal of sFlt-1 from maternal circulation has shown promise in delaying disease progression and improving outcomes. Recently, the FDA approved the first molecular test for preterm preeclampsia (sFlt-1/PlGF ratio) for clinical use in the United States. Measuring serum sFlt-1/PlGF ratio in the acute hospital setting may aid short-term management, particularly regarding step-up or step-down of care, decision to transfer to settings better equipped to manage both the mother and the preterm neonate, appropriate timing of administration of steroids and magnesium sulfate, and in expectant management decisions. The test itself has the potential to save lives. Furthermore, the availability of a molecular test that correlates with adverse outcomes has set the stage for interventional clinical trials testing treatments for this disorder. In this review, we will discuss the role of circulating sFlt-1 and related factors in the pathogenesis of preeclampsia and specifically how this discovery is leading to concrete advances in the care of women with preeclampsia.
Collapse
Affiliation(s)
- Ravi Thadhani
- Woodruff Health Sciences Center, Emory University School of Medicine, Atlanta, Georgia, USA
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Ana Sofia Cerdeira
- Nuffield Department of Women's Health and Reproductive Research, University of Oxford, Oxford, UK
- Fetal Maternal Medicine Unit, Queen Charlotte's and Chelsea Hospital, London, UK
| | - S Ananth Karumanchi
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
17
|
van Hoorn E, Rademaker D, van der Wel A, DeVries J, Franx A, van Rijn B, Kooy A, Siegelaar S, Roseboom T, Ozanne S, Hooijmans C, Painter R. Fetal and post-natal outcomes in offspring after intrauterine metformin exposure: A systematic review and meta-analysis of animal experiments. Diabet Med 2024; 41:e15243. [PMID: 37845186 PMCID: PMC7617357 DOI: 10.1111/dme.15243] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/18/2023]
Abstract
AIMS The impact of maternal metformin use during pregnancy on fetal, infant, childhood and adolescent growth, development, and health remains unclear. Our objective was to systematically review the available evidence from animal experiments on the effects of intrauterine metformin exposure on offspring's anthropometric, cardiovascular and metabolic outcomes. METHODS A systematic search was conducted in PUBMED and EMBASE from inception (searched on 12th April 2023). We extracted original, controlled animal studies that investigated the effects of maternal metformin use during pregnancy on offspring anthropometric, cardiovascular and metabolic measurements. Subsequently, risk of bias was assessed and meta-analyses using the standardized mean difference and a random effects model were conducted for all outcomes containing data from 3 or more studies. Subgroup analyses were planned for species, strain, sex and type of model in the case of 10 comparisons or more per subgroup. RESULTS We included 37 articles (n = 3133 offspring from n = 716 litters, containing n = 51 comparisons) in this review, mostly (95%) on rodent models and 5% pig models. Follow-up of offspring ranged from birth to 2 years of age. Thirty four of the included articles could be included in the meta-analysis. No significant effects in the overall meta-analysis of metformin on any of the anthropometric, cardiovascular and metabolic offspring outcome measures were identified. Between-studies heterogeneity was high, and risk of bias was unclear in most studies as a consequence of poor reporting of essential methodological details. CONCLUSION This systematic review was unable to establish effects of metformin treatment during pregnancy on anthropometric, cardiovascular and metabolic outcomes in non-human offspring. Heterogeneity between studies was high and reporting of methodological details often limited. This highlights a need for additional high-quality research both in humans and model systems to allow firm conclusions to be established. Future research should include focus on the effects of metformin in older offspring age groups, and on outcomes which have gone uninvestigated to date.
Collapse
Affiliation(s)
- E.G.M. van Hoorn
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - D. Rademaker
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
- Department of Obstetrics and Gynecology, Amsterdam University Medical Center location AMC, Amsterdam, The Netherlands
| | - A.W.T. van der Wel
- Department of Obstetrics and Gynecology, Amsterdam University Medical Center location AMC, Amsterdam, The Netherlands
| | - J.H. DeVries
- Department of Internal Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - A. Franx
- Department of Obstetrics and Gynecology Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - B.B. van Rijn
- Department of Obstetrics and Gynecology Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - A. Kooy
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Bethesda Diabetes Research Center, Hoogeveen, The Netherlands
- Department of Internal Medicine, Care Group Treant, Location Bethesda Hoogeveen, Hoogeveen, The Netherlands
| | - S.E. Siegelaar
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam, The Netherlands
| | - T.J. Roseboom
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
- Department of Obstetrics and Gynecology, Amsterdam University Medical Center location AMC, Amsterdam, The Netherlands
- Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - S.E. Ozanne
- Welcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - C.R. Hooijmans
- Department of Anesthesiology, Pain and Palliative Care (Meta Research Team), Radboud University Medical Center, Nijmegen, The Netherlands
| | - R.C. Painter
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
- Department of Obstetrics and Gynecology, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Paschou SA, Shalit A, Gerontiti E, Athanasiadou KI, Kalampokas T, Psaltopoulou T, Lambrinoudaki I, Anastasiou E, Wolffenbuttel BHR, Goulis DG. Efficacy and safety of metformin during pregnancy: an update. Endocrine 2024; 83:259-269. [PMID: 37798604 PMCID: PMC10850184 DOI: 10.1007/s12020-023-03550-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/23/2023] [Indexed: 10/07/2023]
Abstract
During the last decades, gestational diabetes mellitus (GDM) prevalence has been on the rise. While insulin remains the gold standard treatment for GDM, metformin use during pregnancy is controversial. This review aimed to comprehensively assess the available data on the efficacy and safety of metformin during pregnancy, both for the mother and the offspring. Metformin has been validated for maternal efficacy and safety, achieving comparable glycemic control with insulin. Additionally, it reduces maternal weight gain and possibly the occurrence of hypertensive disorders. During the early neonatal period, metformin administration does not increase the risk of congenital anomalies or other major adverse effects, including lower APGAR score at 5 min, neonatal intensive care unit admissions, and respiratory distress syndrome. Several studies have demonstrated a reduction in neonatal hypoglycemia. Metformin has been associated with an increase in preterm births and lower birth weight, although this effect is controversial and depends on the indication for which it was administered. Evidence indicates possible altered fetal programming and predisposition to childhood obesity and metabolic syndrome during adulthood after use of metformin in pregnancy. With critical questions still requiring a final verdict, ongoing research on the field must be conducted.
Collapse
Affiliation(s)
- Stavroula A Paschou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| | - Almog Shalit
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleni Gerontiti
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Kleoniki I Athanasiadou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodoros Kalampokas
- Second Department of Obstetrics and Gynecology, Aretaieion University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodora Psaltopoulou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Irene Lambrinoudaki
- Second Department of Obstetrics and Gynecology, Aretaieion University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Bruce H R Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dimitrios G Goulis
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
19
|
Tarry-Adkins JL, Robinson IG, Pantaleão LC, Armstrong JL, Thackray BD, Holzner LMW, Knapton AE, Virtue S, Jenkins B, Koulman A, Murray AJ, Ozanne SE, Aiken CE. The metabolic response of human trophoblasts derived from term placentas to metformin. Diabetologia 2023; 66:2320-2331. [PMID: 37670017 PMCID: PMC10627909 DOI: 10.1007/s00125-023-05996-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/18/2023] [Indexed: 09/07/2023]
Abstract
AIMS/HYPOTHESIS Metformin is increasingly used therapeutically during pregnancy worldwide, particularly in the treatment of gestational diabetes, which affects a substantial proportion of pregnant women globally. However, the impact on placental metabolism remains unclear. In view of the association between metformin use in pregnancy and decreased birthweight, it is essential to understand how metformin modulates the bioenergetic and anabolic functions of the placenta. METHODS A cohort of 55 placentas delivered by elective Caesarean section at term was collected from consenting participants. Trophoblasts were isolated from the placental samples and treated in vitro with clinically relevant doses of metformin (0.01 mmol/l or 0.1 mmol/l) or vehicle. Respiratory function was assayed using high-resolution respirometry to measure oxygen concentration and calculated [Formula: see text]. Glycolytic rate and glycolytic stress assays were performed using Agilent Seahorse XF assays. Fatty acid uptake and oxidation measurements were conducted using radioisotope-labelled assays. Lipidomic analysis was conducted using LC-MS. Gene expression and protein analysis were performed using RT-PCR and western blotting, respectively. RESULTS Complex I-supported oxidative phosphorylation was lower in metformin-treated trophoblasts (0.01 mmol/l metformin, 61.7% of control, p<0.05; 0.1 mmol/l metformin, 43.1% of control, p<0.001). The proton efflux rate arising from glycolysis under physiological conditions was increased following metformin treatment, up to 23±5% above control conditions following treatment with 0.1 mmol/l metformin (p<0.01). There was a significant increase in triglyceride concentrations in trophoblasts treated with 0.1 mmol/l metformin (p<0.05), particularly those of esters of long-chain polyunsaturated fatty acids. Fatty acid oxidation was reduced by ~50% in trophoblasts treated with 0.1 mmol/l metformin compared with controls (p<0.001), with no difference in uptake between treatment groups. CONCLUSIONS/INTERPRETATION In primary trophoblasts derived from term placentas metformin treatment caused a reduction in oxidative phosphorylation through partial inactivation of complex I and potentially by other mechanisms. Metformin-treated trophoblasts accumulate lipids, particularly long- and very-long-chain polyunsaturated fatty acids. Our findings raise clinically important questions about the balance of risk of metformin use during pregnancy, particularly in situations where the benefits are not clear-cut and alternative therapies are available.
Collapse
Affiliation(s)
- Jane L Tarry-Adkins
- Department of Obstetrics and Gynaecology, the Rosie Hospital and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - India G Robinson
- Department of Obstetrics and Gynaecology, the Rosie Hospital and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Lucas C Pantaleão
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Jenna L Armstrong
- Department of Physiology, Neuroscience and Development, University of Cambridge, Cambridge, UK
| | - Benjamin D Thackray
- Department of Physiology, Neuroscience and Development, University of Cambridge, Cambridge, UK
| | - Lorenz M W Holzner
- Department of Physiology, Neuroscience and Development, University of Cambridge, Cambridge, UK
| | - Alice E Knapton
- Department of Physiology, Neuroscience and Development, University of Cambridge, Cambridge, UK
| | - Sam Virtue
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Benjamin Jenkins
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Albert Koulman
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Andrew J Murray
- Department of Physiology, Neuroscience and Development, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Susan E Ozanne
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Catherine E Aiken
- Department of Obstetrics and Gynaecology, the Rosie Hospital and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK.
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
20
|
Cluver CA, Bergman L, Bergkvist J, Imberg H, Geerts L, Hall DR, Mol BW, Tong S, Walker SP. Impact of fetal growth restriction on pregnancy outcome in women undergoing expectant management for preterm pre-eclampsia. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2023; 62:660-667. [PMID: 37289938 PMCID: PMC10947051 DOI: 10.1002/uog.26282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/18/2023] [Accepted: 05/24/2023] [Indexed: 06/10/2023]
Abstract
OBJECTIVES To assess whether coexisting fetal growth restriction (FGR) influences pregnancy latency among women with preterm pre-eclampsia undergoing expectant management. Secondary outcomes assessed were indication for delivery, mode of delivery and rate of serious adverse maternal and perinatal outcomes. METHODS We conducted a secondary analysis of the Pre-eclampsia Intervention (PIE) and the Pre-eclampsia Intervention 2 (PI2) trial data. These randomized controlled trials evaluated whether esomeprazole and metformin could prolong gestation of women diagnosed with pre-eclampsia between 26 and 32 weeks of gestation undergoing expectant management. Delivery indications were deteriorating maternal or fetal status, or reaching 34 weeks' gestation. FGR (defined by Delphi consensus) at the time of pre-eclampsia diagnosis was examined as a predictor of outcome. Only placebo data from PI2 were included, as the trial showed that metformin use was associated with prolonged gestation. All outcome data were collected prospectively from diagnosis of pre-eclampsia to 6 weeks after the expected due date. RESULTS Of the 202 women included, 92 (45.5%) had FGR at the time of pre-eclampsia diagnosis. Median pregnancy latency was 6.8 days in the FGR group and 15.3 days in the control group (difference 8.5 days; adjusted 0.49-fold change (95% CI, 0.33-0.74); P < 0.001). FGR pregnancies were less likely to reach 34 weeks' gestation (12.0% vs 30.9%; adjusted relative risk (aRR), 0.44 (95% CI, 0.23-0.83)) and more likely to be delivered for suspected fetal compromise (64.1% vs 36.4%; aRR, 1.84 (95% CI, 1.36-2.47)). More women with FGR underwent a prelabor emergency Cesarean section (66.3% vs 43.6%; aRR, 1.56 (95% CI, 1.20-2.03)) and were less likely to have a successful induction of labor (4.3% vs 14.5%; aRR, 0.32 (95% CI, 0.10-1.00)), compared to those without FGR. The rate of maternal complications did not differ significantly between the two groups. FGR was associated with a higher rate of infant death (14.1% vs 4.5%; aRR, 3.26 (95% CI, 1.08-9.81)) and need for intubation and mechanical ventilation (15.2% vs 5.5%; aRR, 2.97 (95% CI, 1.11-7.90)). CONCLUSION FGR is commonly present in women with early preterm pre-eclampsia and outcome is poorer. FGR is associated with shorter pregnancy latency, more emergency Cesarean deliveries, fewer successful inductions and increased rates of neonatal morbidity and mortality. © 2023 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- C. A. Cluver
- Department of Obstetrics and GynaecologyStellenbosch University and Tygerberg HospitalCape TownSouth Africa
- Mercy PerinatalMercy Hospital for WomenMelbourneVIAustralia
- Translational Obstetrics GroupUniversity of MelbourneMelbourneVIAustralia
| | - L. Bergman
- Department of Obstetrics and GynaecologyStellenbosch University and Tygerberg HospitalCape TownSouth Africa
- Department of Obstetrics and GynecologyInstitute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - J. Bergkvist
- Department of Obstetrics and GynecologyInstitute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - H. Imberg
- Statistiska KonsultgruppenGothenburgSweden
- Department of Mathematical SciencesChalmers University of Technology and University of GothenburgGothenburgSweden
| | - L. Geerts
- Department of Obstetrics and GynaecologyStellenbosch University and Tygerberg HospitalCape TownSouth Africa
| | - D. R. Hall
- Department of Obstetrics and GynaecologyStellenbosch University and Tygerberg HospitalCape TownSouth Africa
| | - B. W. Mol
- Department of Obstetrics and Gynaecology, Monash School of MedicineMonash UniversityMelbourneVIAustralia
- Aberdeen Centre for Women's Health Research, Institute of Applied Health Sciences, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenAberdeenUK
| | - S. Tong
- Mercy PerinatalMercy Hospital for WomenMelbourneVIAustralia
- Translational Obstetrics GroupUniversity of MelbourneMelbourneVIAustralia
| | - S. P. Walker
- Mercy PerinatalMercy Hospital for WomenMelbourneVIAustralia
- Translational Obstetrics GroupUniversity of MelbourneMelbourneVIAustralia
| |
Collapse
|
21
|
Grimaldi B, Kohan-Ghadr HR, Halari CD, Nandi P, Kingdom JC, Drewlo S. Rosiglitazone-Mediated Activation of PPARγ Restores HO1 Expression in the Human Preeclamptic Placenta. Hypertension 2023; 80:2386-2396. [PMID: 37702083 PMCID: PMC10581437 DOI: 10.1161/hypertensionaha.123.21645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/14/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND Preeclampsia is a hypertensive disorder of pregnancy characterized by chronic placental ischemia and suppression of proangiogenic proteins, causing oxidative stress, hypertension, and maternal systemic organ damage. The transcription factor, PPARγ (peroxisome proliferator-activated receptor-γ) promotes healthy trophoblast differentiation but is dysregulated in the preeclampsia placenta. Our study identifies the beneficial impact of Rosiglitazone-mediated PPARγ-activation in the stressed preeclampsia placenta. METHODS We used first trimester placentas, preeclamptic and preterm control placentas, and human trophoblast cell lines to study PPARγ activation. RESULTS Induction of PPARγ activates cell growth and antioxidative stress pathways, including the gene, heme oxygenase 1 (Hmox1). Protein expression of both PPARγ and HO1 (heme oxygenase 1) are reduced in preeclamptic placentas, but Rosiglitazone restores HO1 signaling in a PPARγ-dependent manner. CONCLUSIONS Restoring disrupted pathways by PPARγ in preeclampsia offers a potential therapeutic pathway to reverse placental damage, extending pregnancy duration, and reduce maternal sequelae. Future research should aim to understand the full scope of impaired PPARγ signaling in the human placenta and focus on compounds for safe use during pregnancy to prevent severe perinatal morbidity and mortality.
Collapse
Affiliation(s)
- Brooke Grimaldi
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI (B.G., H.-R.K.-G.)
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI (B.G.)
| | - Hamid-Reza Kohan-Ghadr
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI (B.G., H.-R.K.-G.)
| | - Chidambra D. Halari
- Biological Sciences Platform, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Canada (C.D.H., P.N., S.D.)
| | - Pinki Nandi
- Biological Sciences Platform, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Canada (C.D.H., P.N., S.D.)
| | - John C. Kingdom
- Department of Obstetrics and Gynecology, Temerty Faculty of Medicine, University of Toronto, Canada (J.C.K., S.D.)
- Department of Maternal-Fetal Medicine, University of Toronto, Canada (J.C.K.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (J.C.K., S.D.)
| | - Sascha Drewlo
- Department of Obstetrics and Gynecology, Temerty Faculty of Medicine, University of Toronto, Canada (J.C.K., S.D.)
- Biological Sciences Platform, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Canada (C.D.H., P.N., S.D.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (J.C.K., S.D.)
| |
Collapse
|
22
|
Kingdom J, Ashwal E, Lausman A, Liauw J, Soliman N, Figueiro-Filho E, Nash C, Bujold E, Melamed N. Directive clinique n o 442 : Retard de croissance intra-utérin : Dépistage, diagnostic et prise en charge en contexte de grossesse monofœtale. JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2023; 45:102155. [PMID: 37730301 DOI: 10.1016/j.jogc.2023.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
OBJECTIF Le retard de croissance intra-utérin est une complication obstétricale fréquente qui touche jusqu'à 10 % des grossesses dans la population générale et qui est le plus souvent due à une pathologie placentaire sous-jacente. L'objectif de la présente directive clinique est de fournir des déclarations sommaires et des recommandations pour appuyer un protocole clinique de dépistage, diagnostic et prise en charge du retard de croissance intra-utérin pour les grossesses à risque ou atteintes. POPULATION CIBLE Toutes les patientes enceintes menant une grossesse monofœtale. BéNéFICES, RISQUES ET COûTS: La mise en application des recommandations de la présente directive devrait améliorer la compétence des cliniciens quant à la détection du retard de croissance intra-utérin et à la réalisation des interventions indiquées. DONNéES PROBANTES: La littérature publiée a été colligée par des recherches effectuées jusqu'en septembre 2022 dans les bases de données PubMed, Medline, CINAHL et Cochrane Library en utilisant un vocabulaire contrôlé au moyen de termes MeSH pertinents (fetal growth retardation and small for gestational age) et de mots-clés (fetal growth, restriction, growth retardation, IUGR, FGR, low birth weight, small for gestational age, Doppler, placenta, pathology). Seuls les résultats de revues systématiques, d'essais cliniques randomisés ou comparatifs et d'études observationnelles ont été retenus. La littérature grise a été obtenue par des recherches menées dans des sites Web d'organismes s'intéressant à l'évaluation des technologies dans le domaine de la santé et d'organismes connexes, dans des collections de directives cliniques, des registres d'essais cliniques et des sites Web de sociétés de spécialité médicale nationales et internationales. MéTHODES DE VALIDATION: Les auteurs ont évalué la qualité des données probantes et la force des recommandations en utilisant le cadre méthodologique GRADE (Grading of Recommendations Assessment, Development and Evaluation). Voir l'annexe A en ligne (tableau A1 pour les définitions et tableau A2 pour l'interprétation des recommandations fortes et conditionnelles [faibles]). PROFESSIONNELS CONCERNéS: Obstétriciens, médecins de famille, infirmières, sages-femmes, spécialistes en médecine fœto-maternelle, radiologistes et autres professionnels de la santé qui prodiguent des soins aux patientes enceintes. RéSUMé POUR TWITTER: Mise à jour de la directive sur le dépistage, le diagnostic et la prise en charge du retard de croissance intra-utérin pour les grossesses à risque ou atteintes. DÉCLARATIONS SOMMAIRES: RECOMMANDATIONS: Prédiction du retard de croissance intra-utérin Prévention du retard de croissance intra-utérin Détection du retard de croissance intra-utérin Examens en cas de retard de croissance intra-utérin soupçonné Prise en charge du retard de croissance intra-utérin précoce Prise en charge du retard de croissance intra-utérin tardif Prise en charge du post-partum et consultations préconception.
Collapse
|
23
|
Kingdom J, Ashwal E, Lausman A, Liauw J, Soliman N, Figueiro-Filho E, Nash C, Bujold E, Melamed N. Guideline No. 442: Fetal Growth Restriction: Screening, Diagnosis, and Management in Singleton Pregnancies. JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2023; 45:102154. [PMID: 37730302 DOI: 10.1016/j.jogc.2023.05.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
OBJECTIVE Fetal growth restriction is a common obstetrical complication that affects up to 10% of pregnancies in the general population and is most commonly due to underlying placental diseases. The purpose of this guideline is to provide summary statements and recommendations to support a clinical framework for effective screening, diagnosis, and management of pregnancies that are either at risk of or affected by fetal growth restriction. TARGET POPULATION All pregnant patients with a singleton pregnancy. BENEFITS, HARMS, AND COSTS Implementation of the recommendations in this guideline should increase clinician competency to detect fetal growth restriction and provide appropriate interventions. EVIDENCE Published literature in English was retrieved through searches of PubMed or MEDLINE, CINAHL, and The Cochrane Library through to September 2022 using appropriate controlled vocabulary via MeSH terms (fetal growth retardation and small for gestational age) and key words (fetal growth, restriction, growth retardation, IUGR, FGR, low birth weight, small for gestational age, Doppler, placenta, pathology). Results were restricted to systematic reviews, randomized controlled trials/controlled clinical trials, and observational studies. Grey literature was identified through searching the websites of health technology assessment and health technology-related agencies, clinical practice guideline collections, clinical trial registries, and national and international medical specialty societies. VALIDATION METHODS The authors rated the quality of evidence and strength of recommendations using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach. See online Appendix A (Table A1 for definitions and Table A2 for interpretations of strong and conditional [weak] recommendations). INTENDED AUDIENCE Obstetricians, family physicians, nurses, midwives, maternal-fetal medicine specialists, radiologists, and other health care providers who care for pregnant patients. TWEETABLE ABSTRACT Updated guidelines on screening, diagnosis, and management of pregnancies at risk of or affected by FGR. SUMMARY STATEMENTS RECOMMENDATIONS: Prediction of FGR Prevention of FGR Detection of FGR Investigations in Pregnancies with Suspected Fetal Growth Restriction Management of Early-Onset Fetal Growth Restriction Management of Late-Onset FGR Postpartum management and preconception counselling.
Collapse
|
24
|
Tocci V, Mirabelli M, Salatino A, Sicilia L, Giuliano S, Brunetti FS, Chiefari E, De Sarro G, Foti DP, Brunetti A. Metformin in Gestational Diabetes Mellitus: To Use or Not to Use, That Is the Question. Pharmaceuticals (Basel) 2023; 16:1318. [PMID: 37765126 PMCID: PMC10537239 DOI: 10.3390/ph16091318] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
In recent years, there has been a dramatic increase in the number of pregnancies complicated by gestational diabetes mellitus (GDM). GDM occurs when maternal insulin resistance develops and/or progresses during gestation, and it is not compensated by a rise in maternal insulin secretion. If not properly managed, this condition can cause serious short-term and long-term problems for both mother and child. Lifestyle changes are the first line of treatment for GDM, but if ineffective, insulin injections are the recommended pharmacological treatment choice. Some guidance authorities and scientific societies have proposed the use of metformin as an alternative pharmacological option for treating GDM, but there is not yet a unanimous consensus on this. Although the use of metformin appears to be safe for the mother, concerns remain about its long-term metabolic effects on the child that is exposed in utero to the drug, given that metformin, contrary to insulin, crosses the placenta. This review article describes the existing lines of evidence about the use of metformin in pregnancies complicated by GDM, in order to clarify its potential benefits and limits, and to help clinicians make decisions about who could benefit most from this drug treatment.
Collapse
Affiliation(s)
- Vera Tocci
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (V.T.); (M.M.)
- Operative Unit of Endocrinology, Diabetes in Pregnancy Ambulatory Care Center, Renato Dulbecco University Hospital, 88100 Catanzaro, Italy
| | - Maria Mirabelli
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (V.T.); (M.M.)
- Operative Unit of Endocrinology, Diabetes in Pregnancy Ambulatory Care Center, Renato Dulbecco University Hospital, 88100 Catanzaro, Italy
| | - Alessandro Salatino
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (V.T.); (M.M.)
| | - Luciana Sicilia
- Operative Unit of Endocrinology, Diabetes in Pregnancy Ambulatory Care Center, Renato Dulbecco University Hospital, 88100 Catanzaro, Italy
| | - Stefania Giuliano
- Operative Unit of Endocrinology, Diabetes in Pregnancy Ambulatory Care Center, Renato Dulbecco University Hospital, 88100 Catanzaro, Italy
| | - Francesco S. Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (V.T.); (M.M.)
| | - Eusebio Chiefari
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (V.T.); (M.M.)
| | - Giovambattista De Sarro
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (V.T.); (M.M.)
| | - Daniela P. Foti
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy;
| | - Antonio Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (V.T.); (M.M.)
- Operative Unit of Endocrinology, Diabetes in Pregnancy Ambulatory Care Center, Renato Dulbecco University Hospital, 88100 Catanzaro, Italy
| |
Collapse
|
25
|
Casey H, Dennehy N, Fraser A, Lees C, McEniery C, Scott K, Wilkinson I, Delles C. Placental syndromes and maternal cardiovascular health. Clin Sci (Lond) 2023; 137:1211-1224. [PMID: 37606085 PMCID: PMC10447226 DOI: 10.1042/cs20211130] [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: 02/05/2023] [Revised: 07/16/2023] [Accepted: 08/07/2023] [Indexed: 08/23/2023]
Abstract
The placental syndromes gestational hypertension, preeclampsia and intrauterine growth restriction are associated with an increased cardiovascular risk to the mother later in life. In this review, we argue that a woman's pre-conception cardiovascular health drives both the development of placental syndromes and long-term cardiovascular risk but acknowledge that placental syndromes can also contribute to future cardiovascular risk independent of pre-conception health. We describe how preclinical studies in models of preeclampsia inform our understanding of the links with later cardiovascular disease, and how current pre-pregnancy studies may explain relative contributions of both pre-conception factors and the occurrence of placental syndromes to long-term cardiovascular disease.
Collapse
Affiliation(s)
- Helen Casey
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, Scotland, U.K
| | - Natalie Dennehy
- Chelsea and Westminster NHS Foundation Trust, London, England, U.K
| | - Abigail Fraser
- Department of Population Health Sciences, Bristol Medical School, and the MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, U.K
| | - Christoph Lees
- Chelsea and Westminster NHS Foundation Trust, London, England, U.K
| | - Carmel M. McEniery
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, England, U.K
| | - Kayley Scott
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, Scotland, U.K
| | - Ian B. Wilkinson
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, England, U.K
| | - Christian Delles
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, Scotland, U.K
| |
Collapse
|
26
|
Ryssdal M, Vanky E, Stokkeland LMT, Jarmund AH, Steinkjer B, Løvvik TS, Madssen TS, Iversen AC, Giskeødegård GF. Immunomodulatory Effects of Metformin Treatment in Pregnant Women With PCOS. J Clin Endocrinol Metab 2023; 108:e743-e753. [PMID: 36916886 PMCID: PMC10438881 DOI: 10.1210/clinem/dgad145] [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: 09/06/2022] [Revised: 03/05/2023] [Accepted: 03/07/2023] [Indexed: 03/15/2023]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS) is a common endocrine disorder associated with low-grade systemic inflammation and increased risk of pregnancy complications. Metformin treatment reduces the risk of late miscarriage and preterm birth in pregnant women with PCOS. Whether the protective effect of metformin involves immunological changes has not been determined. OBJECTIVE To investigate the effect of metformin on the maternal immunological status in women with PCOS. METHODS A post-hoc analysis was performed of two randomized controlled trials, PregMet and PregMet2, including longitudinal maternal serum samples from 615 women with PCOS. Women were randomized to metformin or placebo from first trimester to delivery. Twenty-two cytokines and C-reactive protein were measured in serum sampled at gestational weeks 5 to 12, 19, 32, and 36. RESULTS Metformin treatment was associated with higher serum levels of several multifunctional cytokines throughout pregnancy, with the strongest effect on eotaxin (P < .001), interleukin-17 (P = .03), and basic fibroblast growth factor (P = .04). Assessment of the combined cytokine development confirmed the impact of metformin on half of the 22 cytokines. The immunomodulating effect of metformin was more potent in normal weight and overweight women than in obese women. Moreover, normoandrogenic women had the strongest effect of metformin in early pregnancy, whereas hyperandrogenic women presented increasing effect throughout pregnancy. CONCLUSION It appears that metformin has immunomodulating rather than anti-inflammatory properties in pregnancy. Its effect on the serum levels of many multifunctional cytokines demonstrates robust, persisting, and body mass-dependent immune mobilization in pregnant women with PCOS.
Collapse
Affiliation(s)
- Mariell Ryssdal
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
- Centre of Molecular Inflammation Research (CEMIR), Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Eszter Vanky
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
- Department of Obstetrics and Gynecology, St. Olavs Hospital, Trondheim University Hospital, 7006 Trondheim, Norway
| | - Live Marie T Stokkeland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
- Centre of Molecular Inflammation Research (CEMIR), Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Anders Hagen Jarmund
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
- Centre of Molecular Inflammation Research (CEMIR), Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Bjørg Steinkjer
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
- Centre of Molecular Inflammation Research (CEMIR), Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Tone Shetelig Løvvik
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
- Department of Obstetrics and Gynecology, St. Olavs Hospital, Trondheim University Hospital, 7006 Trondheim, Norway
| | - Torfinn Støve Madssen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Ann-Charlotte Iversen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
- Centre of Molecular Inflammation Research (CEMIR), Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Department of Obstetrics and Gynecology, St. Olavs Hospital, Trondheim University Hospital, 7006 Trondheim, Norway
| | - Guro F Giskeødegård
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| |
Collapse
|
27
|
Sakowicz A, Bralewska M, Rybak-Krzyszkowska M, Grzesiak M, Pietrucha T. New Ideas for the Prevention and Treatment of Preeclampsia and Their Molecular Inspirations. Int J Mol Sci 2023; 24:12100. [PMID: 37569476 PMCID: PMC10418829 DOI: 10.3390/ijms241512100] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Preeclampsia (PE) is a pregnancy-specific disorder affecting 4-10% of all expectant women. It greatly increases the risk of maternal and foetal death. Although the main symptoms generally appear after week 20 of gestation, scientific studies indicate that the mechanism underpinning PE is initiated at the beginning of gestation. It is known that the pathomechanism of preeclampsia is strongly related to inflammation and oxidative stress, which influence placentation and provoke endothelial dysfunction in the mother. However, as of yet, no "key players" regulating all these processes have been discovered. This might be why current therapeutic strategies intended for prevention or treatment are not fully effective, and the only effective method to stop the disease is the premature induction of delivery, mostly by caesarean section. Therefore, there is a need for further research into new pharmacological strategies for the treatment and prevention of preeclampsia. This review presents new preventive methods and therapies for PE not yet recommended by obstetrical and gynaecological societies. As many of these therapies are in preclinical studies or under evaluation in clinical trials, this paper reports the molecular targets of the tested agents or methods.
Collapse
Affiliation(s)
- Agata Sakowicz
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland; (M.B.); (T.P.)
| | - Michalina Bralewska
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland; (M.B.); (T.P.)
| | - Magda Rybak-Krzyszkowska
- Department of Obstetrics and Perinatology, University Hospital in Krakow, 31-501 Krakow, Poland;
| | - Mariusz Grzesiak
- Department of Perinatology, Obstetrics and Gynecology, Polish Mother’s Memorial Hospital-Research Institute in Lodz, 93-338 Lodz, Poland;
- Department of Gynecology and Obstetrics, Medical University of Lodz, 93-338 Lodz, Poland
| | - Tadeusz Pietrucha
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland; (M.B.); (T.P.)
| |
Collapse
|
28
|
Tabacco S, Ambrosii S, Polsinelli V, Fantasia I, D’Alfonso A, Ludovisi M, Cecconi S, Guido M. Pre-Eclampsia: From Etiology and Molecular Mechanisms to Clinical Tools-A Review of the Literature. Curr Issues Mol Biol 2023; 45:6202-6215. [PMID: 37623210 PMCID: PMC10453909 DOI: 10.3390/cimb45080391] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/14/2023] [Accepted: 07/20/2023] [Indexed: 08/26/2023] Open
Abstract
Pre-eclampsia is a severe pregnancy-related complication that manifests as a syndrome with multisystem involvement and damage. It has significantly grown in frequency during the past 30 years and could be considered as one of the major causes of maternal and fetal morbidity and mortality. However, the specific etiology and molecular mechanisms of pre-eclampsia are still poorly known and could have a variety of causes, such as altered angiogenesis, inflammations, maternal infections, obesity, metabolic disorders, gestational diabetes, and autoimmune diseases. Perhaps the most promising area under investigation is the imbalance of maternal angiogenic factors and its effects on vascular function, though studies in placental oxidative stress and maternal immune response have demonstrated intriguing findings. However, to determine the relative importance of each cause and the impact of actions aiming to significantly reduce the incidence of this illness, more research is needed. Moreover, it is necessary to better understand the etiologies of each subtype of pre-eclampsia as well as the pathophysiology of other major obstetrical syndromes to identify a clinical tool able to recognize patients at risk of pre-eclampsia early.
Collapse
Affiliation(s)
- Sara Tabacco
- Unit of Obstetrics and Gynecology, San Salvatore Hospital, 67100 L’Aquila, Italy
| | - Silvia Ambrosii
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Valentina Polsinelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Ilaria Fantasia
- Unit of Obstetrics and Gynecology, San Salvatore Hospital, 67100 L’Aquila, Italy
| | - Angela D’Alfonso
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Manuela Ludovisi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Sandra Cecconi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Maurizio Guido
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| |
Collapse
|
29
|
Abstract
Hypertensive disorders of pregnancy (HDP) are one of the most commonly occurring complications of pregnancy and include chronic hypertension, gestational hypertension, and pre-eclampsia. New developments in early pregnancy screening to identify women at high risk for pre-eclampsia combined with targeted aspirin prophylaxis could greatly reduce the number of affected pregnancies. Furthermore, recent advances in the diagnosis of pre-eclampsia, such as placental growth factor based testing, have been shown to improve the identification of those pregnancies at highest risk of severe complications. Evidence from trials has refined the target blood pressure and timing of delivery to manage chronic hypertension and pre-eclampsia with non-severe features, respectively. Importantly, a wealth of epidemiological data now links HDP to future cardiovascular disease and diabetes decades after an affected pregnancy. This review discusses the current guidelines and research data on the prevention, diagnosis, management, and postnatal follow-up of HDP. It also discusses the gap in knowledge regarding the long term risks for cardiovascular disease following HDP and illustrates the importance of improving adherence to postnatal guidelines to monitor hypertension and the need for more research focused on primary prevention of future cardiovascular disease in women identified as being at high risk because of HDP.
Collapse
Affiliation(s)
- Pensée Wu
- School of Medicine, Keele University, Newcastle-under-Lyme, UK
- Academic Department of Obstetrics and Gynaecology, University Hospital of North Midlands, Stoke-on-Trent, UK
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | - Jenny E Myers
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK
| |
Collapse
|
30
|
Menkhorst E, Santos LL, Zhou W, Yang G, Winship AL, Rainczuk KE, Nguyen P, Zhang JG, Moore P, Williams M, Lê Cao KA, Mansell A, Dimitriadis E. IL11 activates the placental inflammasome to drive preeclampsia. Front Immunol 2023; 14:1175926. [PMID: 37292200 PMCID: PMC10244672 DOI: 10.3389/fimmu.2023.1175926] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/09/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction Preeclampsia is a life-threatening disorder of pregnancy unique to humans. Interleukin (IL)11 is elevated in serum from pregnancies that subsequently develop early-onset preeclampsia and pharmacological elevation of IL11 in pregnant mice causes the development of early-onset preeclampsia-like features (hypertension, proteinuria, and fetal growth restriction). However, the mechanism by which IL11 drives preeclampsia is unknown. Method Pregnant mice were administered PEGylated (PEG)IL11 or control (PEG) from embryonic day (E)10-16 and the effect on inflammasome activation, systolic blood pressure (during gestation and at 50/90 days post-natal), placental development, and fetal/post-natal pup growth measured. RNAseq analysis was performed on E13 placenta. Human 1st trimester placental villi were treated with IL11 and the effect on inflammasome activation and pyroptosis identified by immunohistochemistry and ELISA. Result PEGIL11 activated the placental inflammasome causing inflammation, fibrosis, and acute and chronic hypertension in wild-type mice. Global and placental-specific loss of the inflammasome adaptor protein Asc and global loss of the Nlrp3 sensor protein prevented PEGIL11-induced fibrosis and hypertension in mice but did not prevent PEGIL11-induced fetal growth restriction or stillbirths. RNA-sequencing and histology identified that PEGIL11 inhibited trophoblast differentiation towards spongiotrophoblast and syncytiotrophoblast lineages in mice and extravillous trophoblast lineages in human placental villi. Discussion Inhibition of ASC/NLRP3 inflammasome activity could prevent IL11-induced inflammation and fibrosis in various disease states including preeclampsia.
Collapse
Affiliation(s)
- Ellen Menkhorst
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC, Australia
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Leilani L. Santos
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC, Australia
| | - Wei Zhou
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC, Australia
| | - Guannan Yang
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC, Australia
- Department of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, Australia
| | - Amy L. Winship
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Katarzyna E. Rainczuk
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Philana Nguyen
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC, Australia
| | - Jian-Guo Zhang
- Walter and Eliza Hall Institute, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Paddy Moore
- Abortion and Contraception, Royal Women’s Hospital, Parkville, VIC, Australia
| | - Michelle Williams
- Biomedical Animal Facility, The University of Melbourne, Parkville, VIC, Australia
| | - Kim-Anh Lê Cao
- Department of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, Australia
| | - Ashley Mansell
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Evdokia Dimitriadis
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC, Australia
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
31
|
Vatish M, Powys VR, Cerdeira AS. Novel therapeutic and diagnostic approaches for preeclampsia. Curr Opin Nephrol Hypertens 2023; 32:124-133. [PMID: 36683536 DOI: 10.1097/mnh.0000000000000870] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE OF REVIEW This review will summarize recent findings relating to the diagnostic approach to preeclampsia and current avenues of research aimed at modifying the underlying disease process. RECENT FINDINGS Growing international consensus supports a broad preeclampsia definition that incorporates maternal end-organ and uteroplacental dysfunction. Recent evidence demonstrates that this definition better identifies women and babies at risk of adverse outcomes compared to the traditional definition of hypertension and proteinuria. Multiple studies have demonstrated the usefulness and cost-effectiveness of angiogenic biomarkers such as soluble fms-like tyrosine kinase-1 and placental growth factor as a clinical adjunct to diagnose and predict severity of preeclampsia associated outcomes. Current novel therapeutic approaches to preeclampsia target pathogenic pathways (e.g. antiangiogenesis) or downstream effects such as oxidative stress and nitric oxide. Recent findings relating to these promising candidates are discussed. Multicenter clinical trials are needed to evaluate their effectiveness and ability to improve fetal and maternal outcomes. SUMMARY We provide an updated framework of the current approaches to define and diagnose preeclampsia. Disease modifying therapies (in particular, targeting the angiogenic pathway) are being developed for the first time and promise to revolutionize the way we manage preeclampsia.
Collapse
Affiliation(s)
- Manu Vatish
- Nuffield Department of Women's Health and Reproductive Research, University of Oxford, Oxford
| | | | - Ana Sofia Cerdeira
- Nuffield Department of Women's Health and Reproductive Research, University of Oxford, Oxford
| |
Collapse
|
32
|
Placental Mitochondrial Function and Dysfunction in Preeclampsia. Int J Mol Sci 2023; 24:ijms24044177. [PMID: 36835587 PMCID: PMC9963167 DOI: 10.3390/ijms24044177] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
The placenta is a vital organ of pregnancy, regulating adaptation to pregnancy, gestational parent/fetal exchange, and ultimately, fetal development and growth. Not surprisingly, in cases of placental dysfunction-where aspects of placental development or function become compromised-adverse pregnancy outcomes can result. One common placenta-mediated disorder of pregnancy is preeclampsia (PE), a hypertensive disorder of pregnancy with a highly heterogeneous clinical presentation. The wide array of clinical characteristics observed in pregnant individuals and neonates of a PE pregnancy are likely the result of distinct forms of placental pathology underlying the PE diagnosis, explaining why no one common intervention has proven effective in the prevention or treatment of PE. The historical paradigm of placental pathology in PE highlights an important role for utero-placental malperfusion, placental hypoxia and oxidative stress, and a critical role for placental mitochondrial dysfunction in the pathogenesis and progression of the disease. In the current review, the evidence of placental mitochondrial dysfunction in the context of PE will be summarized, highlighting how altered mitochondrial function may be a common feature across distinct PE subtypes. Further, advances in this field of study and therapeutic targeting of mitochondria as a promising intervention for PE will be discussed.
Collapse
|
33
|
Du Y, Zhu YJ, Zhou YX, Ding J, Liu JY. Metformin in therapeutic applications in human diseases: its mechanism of action and clinical study. MOLECULAR BIOMEDICINE 2022; 3:41. [PMID: 36484892 PMCID: PMC9733765 DOI: 10.1186/s43556-022-00108-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/18/2022] [Indexed: 12/13/2022] Open
Abstract
Metformin, a biguanide drug, is the most commonly used first-line medication for type 2 diabetes mellites due to its outstanding glucose-lowering ability. After oral administration of 1 g, metformin peaked plasma concentration of approximately 20-30 μM in 3 h, and then it mainly accumulated in the gastrointestinal tract, liver and kidney. Substantial studies have indicated that metformin exerts its beneficial or deleterious effect by multiple mechanisms, apart from AMPK-dependent mechanism, also including several AMPK-independent mechanisms, such as restoring of redox balance, affecting mitochondrial function, modulating gut microbiome and regulating several other signals, such as FBP1, PP2A, FGF21, SIRT1 and mTOR. On the basis of these multiple mechanisms, researchers tried to repurpose this old drug and further explored the possible indications and adverse effects of metformin. Through investigating with clinical studies, researchers concluded that in addition to decreasing cardiovascular events and anti-obesity, metformin is also beneficial for neurodegenerative disease, polycystic ovary syndrome, aging, cancer and COVID-19, however, it also induces some adverse effects, such as gastrointestinal complaints, lactic acidosis, vitamin B12 deficiency, neurodegenerative disease and offspring impairment. Of note, the dose of metformin used in most studies is much higher than its clinically relevant dose, which may cast doubt on the actual effects of metformin on these disease in the clinic. This review summarizes these research developments on the mechanism of action and clinical evidence of metformin and discusses its therapeutic potential and clinical safety.
Collapse
Affiliation(s)
- Yang Du
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Ya-Juan Zhu
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yi-Xin Zhou
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Jing Ding
- grid.54549.390000 0004 0369 4060Department of Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan China
| | - Ji-Yan Liu
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Schoonejans JM, Blackmore HL, Ashmore TJ, Pantaleão LC, Pellegrini Pisani L, Dearden L, Tadross JA, Aiken CE, Fernandez-Twinn DS, Ozanne SE. Sex-specific effects of maternal metformin intervention during glucose-intolerant obese pregnancy on body composition and metabolic health in aged mouse offspring. Diabetologia 2022; 65:2132-2145. [PMID: 36112170 PMCID: PMC9630251 DOI: 10.1007/s00125-022-05789-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/15/2022] [Indexed: 02/02/2023]
Abstract
AIMS/HYPOTHESIS Metformin is increasingly used to treat gestational diabetes (GDM) and pregnancies complicated by pregestational type 2 diabetes or polycystic ovary syndrome but data regarding long-term offspring outcome are lacking in both human studies and animal models. Using a mouse model, this study investigated the effects of maternal metformin intervention during obese glucose-intolerant pregnancy on adiposity, hepatic steatosis and markers of metabolic health of male and female offspring up to the age of 12 months. METHODS C57BL/6J female mice were weaned onto either a control diet (Con) or, to induce pre-conception obesity, an obesogenic diet (Ob). The respective diets were maintained throughout pregnancy and lactation. These obese dams were then randomised to the untreated group or to receive 300 mg/kg oral metformin hydrochloride treatment (Ob-Met) daily during pregnancy. In male and female offspring, body weights and body composition were measured from 1 month until 12 months of age, when serum and tissues were collected for investigation of adipocyte cellularity (histology), adipose tissue inflammation (histology and quantitative RT-PCR), and hepatic steatosis and fibrosis (histochemistry and modified Folch assay). RESULTS At 12 months of age, male Ob and Ob-Met offspring showed increased adiposity, adipocyte hypertrophy, elevated expression of proinflammatory genes, hyperleptinaemia and hepatic lipid accumulation compared with Con offspring. Male Ob-Met offspring failed to show hyperplasia between 8 weeks and 12 months, indicative of restricted adipose tissue expansion, resulting in increased immune cell infiltration and ectopic lipid deposition. Female Ob offspring were relatively protected from these phenotypes but Ob-Met female offspring showed increased adiposity, adipose tissue inflammation, hepatic lipid accumulation, hyperleptinaemia and hyperinsulinaemia compared with Con female offspring. CONCLUSIONS/INTERPRETATION Maternal metformin treatment of obese dams increased offspring metabolic risk factors in a sex- and age-dependent manner. These observations highlight the importance of following up offspring of both sexes beyond early adulthood after interventions during pregnancy. Our findings illustrate the complexity of balancing short-term benefits to mother and child vs any potential long-term metabolic effects on the offspring when prescribing therapeutic agents that cross the placenta.
Collapse
Affiliation(s)
- Josca M Schoonejans
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK.
- Department of Women and Children's Health, King's College London, London, UK.
| | - Heather L Blackmore
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Thomas J Ashmore
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Lucas C Pantaleão
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Luciana Pellegrini Pisani
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
- Department of Bioscience, Laboratory of Nutrition and Endocrine Physiology, Federal University of São Paulo, Santos, Brazil
| | - Laura Dearden
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - John A Tadross
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Catherine E Aiken
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
- Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge, UK
| | - Denise S Fernandez-Twinn
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Susan E Ozanne
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
35
|
Andres F, Hannan NJ, Walker SP, MacDonald TM, Wong GP, Murphy C, Cannon P, Kandel M, Masci J, Nguyen TV, Abboud A, Idzes D, Kyritsis V, Pritchard N, Tong S, Kaitu'u-Lino TJ. Endothelial protein C receptor is increased in preterm preeclampsia and fetal growth restriction. FASEB J 2022; 36:e22651. [PMID: 36394528 DOI: 10.1096/fj.202201150r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/11/2022] [Accepted: 10/31/2022] [Indexed: 11/18/2022]
Abstract
Placental dysfunction is the leading cause of both preeclampsia and fetal growth restriction. This study aimed to characterize endothelial protein C receptor (EPCR) in preterm preeclampsia, term preeclampsia, and fetal growth restriction (defined by delivery of a small for gestational age [SGA] infant [<10% birthweight centile]) and examine its regulation in primary syncytiotrophoblast. Placental EPCR mRNA and protein were significantly increased in patients with preterm preeclampsia (<34 weeks gestation) compared to gestation-matched controls (p < .0001). In the plasma, EPCR was also significantly elevated (p = .01) in established preterm preeclampsia while its substrate, protein C (PC) was significantly reduced (p = .0083). Placentas from preterm small for gestational age (SGA) cases, had elevated EPCR mRNA expression (p < .0001) relative to controls. At 36 weeks, no significant changes in plasma EPCR were detected in samples from patients destined to develop preeclampsia or deliver an SGA infant at term. In terms of syncytiotrophoblast, hypoxia significantly increased EPCR mRNA expression (p = .008), but Tumor Necrosis Factor Alpha (TNF-α) decreased EPCR mRNA. Interleukin-6 (IL-6) had no significant effect on EPCR mRNA expression. When isolated syncytiotrophoblast was treated with metformin under hypoxia (1% O2 ) or normoxia (8% O2 ), EPCR mRNA expression was significantly reduced (p = .008) relative to control. In conclusion, EPCR is markedly elevated in the placenta and the circulation of patients with established preterm preeclampsia and placental increases may be associated with hypoxia. Additionally, fetal growth-restricted pregnancies (as defined by the delivery of an SGA infant) also demonstrated elevated placental EPCR.
Collapse
Affiliation(s)
- Faith Andres
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, Victoria, Australia.,Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Natalie J Hannan
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia.,Therapeutics Discovery and Vascular Function Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, Victoria, Australia
| | - Susan P Walker
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, Victoria, Australia.,Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Teresa M MacDonald
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, Victoria, Australia.,Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Georgia P Wong
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, Victoria, Australia.,Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Ciara Murphy
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, Victoria, Australia.,Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Ping Cannon
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, Victoria, Australia.,Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Manju Kandel
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, Victoria, Australia.,Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Joshua Masci
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, Victoria, Australia.,Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Tuong-Vi Nguyen
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, Victoria, Australia.,Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Alison Abboud
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Danica Idzes
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Valerie Kyritsis
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Natasha Pritchard
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, Victoria, Australia.,Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Stephen Tong
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, Victoria, Australia.,Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, Victoria, Australia.,Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| |
Collapse
|
36
|
Kadife E, Hannan N, Harper A, Binder N, Beard S, Brownfoot FC. Hydroxychloroquine reduces soluble Flt-1 secretion from human cytotrophoblast, but does not mitigate markers of endothelial dysfunction in vitro. PLoS One 2022; 17:e0271560. [PMID: 36417467 PMCID: PMC9683551 DOI: 10.1371/journal.pone.0271560] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 07/04/2022] [Indexed: 11/26/2022] Open
Abstract
Preeclampsia is a multi-system disease that can have severe, even fatal implications for the mother and fetus. Abnormal placentation can lead to ischaemic tissue injury and placental inflammation. In turn, the placenta releases anti-angiogenic factors into the maternal circulation. These systemically act to neutralise angiogenic factors causing endothelial dysfunction causing preeclampsia. Hydroxychloroquine is an immune modulating drug that is considered safe in pregnancy. There is epidemiological evidence suggesting it may reduce the risk of preeclampsia. Here, we examined the effects hydroxychloroquine on the production and secretion of sFlt-1, soluble endoglin (sENG), placental growth factor (PlGF) and vascular endothelial growth factor (VEGF) in primary human placenta, cytotrophoblasts and umbilical vein endothelial cells (endothelial cell model). Hydroxychloroquine treatment decreased mRNA expression of two sFlt-1 isoforms and its protein secretion. sENG was not reduced. Hydroxychloroquine treatment increased secretion of pro-angiogenic factor PIGF from endothelial cells. It did not significantly reduce the expression of the endothelial cell inflammation marker, ET-1, and inflammation induced expression of the adhesion molecule, VCAM. Hydroxychloroquine could not overcome leukocyte adhesion to endothelial cells. Hydroxychloroquine mitigates features of preeclampsia, but it does not reduce key markers of endothelial dysfunction.
Collapse
Affiliation(s)
- Elif Kadife
- Department of Obstetrics and Gynaecology, Obstetric Diagnostics and Therapeutics Group, University of Melbourne, Melbourne, Victoria, Australia
- Mercy Perinatal, Mercy Hospital for Women, Melbourne, Victoria, Australia
| | - Natalie Hannan
- Mercy Perinatal, Mercy Hospital for Women, Melbourne, Victoria, Australia
- Department of Obstetrics and Gynaecology, Therapeutics Discovery and Vascular Function in Pregnancy, University of Melbourne, Melbourne, Victoria, Australia
| | - Alesia Harper
- Department of Obstetrics and Gynaecology, Obstetric Diagnostics and Therapeutics Group, University of Melbourne, Melbourne, Victoria, Australia
- Mercy Perinatal, Mercy Hospital for Women, Melbourne, Victoria, Australia
| | - Natalie Binder
- Mercy Perinatal, Mercy Hospital for Women, Melbourne, Victoria, Australia
- Department of Obstetrics and Gynaecology, Therapeutics Discovery and Vascular Function in Pregnancy, University of Melbourne, Melbourne, Victoria, Australia
| | - Sally Beard
- Mercy Perinatal, Mercy Hospital for Women, Melbourne, Victoria, Australia
- Department of Obstetrics and Gynaecology, Therapeutics Discovery and Vascular Function in Pregnancy, University of Melbourne, Melbourne, Victoria, Australia
| | - Fiona C. Brownfoot
- Department of Obstetrics and Gynaecology, Obstetric Diagnostics and Therapeutics Group, University of Melbourne, Melbourne, Victoria, Australia
- Mercy Perinatal, Mercy Hospital for Women, Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
37
|
Mcdougall AR, Tuttle A, Goldstein M, Ammerdorffer A, Gülmezoglu AM, Vogel JP. Target product profiles for novel medicines to prevent and treat preeclampsia: An expert consensus. PLOS GLOBAL PUBLIC HEALTH 2022; 2:e0001260. [PMID: 36962694 PMCID: PMC10021561 DOI: 10.1371/journal.pgph.0001260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/23/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Preeclampsia and eclampsia are a leading cause of global maternal and newborn mortality. Currently, there are few effective medicines that can prevent or treat preeclampsia. Target Product Profiles (TPPs) are important tools for driving new product development by specifying upfront the characteristics that new products should take. Considering the lack of investment and innovation around new medicines for obstetric conditions, we aimed to develop two new TPPs for medicines to prevent and treat preeclampsia. METHODS AND FINDINGS We used a multi-methods approach comprised of a literature review, stakeholder interviews, online survey, and public consultation. Following an initial literature review, diverse stakeholders (clinical practice, research, academia, international organizations, funders, consumer representatives) were invited for in-depth interviews and an online international survey, as well as public consultation on draft TPPs. The level of stakeholder agreement with TPPs was assessed, and findings from interviews were synthesised to inform the final TPPs. We performed 23 stakeholder interviews and received 46 survey responses. A high level of agreement was observed in survey results, with 89% of TPP variables reaching consensus (75% agree or strongly agree). Points of discussion were raised around the target population for preeclampsia prevention and treatment, as well as the acceptability of cold-chain storage and routes of administration. CONCLUSION There is consensus within the maternal health research community on the parameters that new medicines for preeclampsia prevention and treatment must achieve to meet real-world health needs. These TPPs provide necessary guidance to spur interest, innovation and investment in the development of new medicines to prevent and treat preeclampsia.
Collapse
Affiliation(s)
- Annie Ra Mcdougall
- Maternal, Child and Adolescent Health Program, Burnet Institute, Melbourne, Australia
| | | | | | | | | | - Joshua P. Vogel
- Maternal, Child and Adolescent Health Program, Burnet Institute, Melbourne, Australia
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| |
Collapse
|
38
|
McDougall ARA, Hastie R, Goldstein M, Tuttle A, Tong S, Ammerdorffer A, Gülmezoglu AM, Vogel JP. Systematic evaluation of the pre-eclampsia drugs, dietary supplements and biologicals pipeline using target product profiles. BMC Med 2022; 20:393. [PMID: 36329468 PMCID: PMC9635102 DOI: 10.1186/s12916-022-02582-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/26/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The Accelerating Innovation for Mothers (AIM) project established a database of candidate medicines in research and development (R&D) between 2000 and 2021 for five pregnancy-related conditions, including pre-eclampsia. In parallel, we published target product profiles (TPPs) that describe optimal characteristics of medicines for use in preventing/treating pre-eclampsia. The study objective was to use systematic double screening and extraction to identify all candidate medicines being investigated for pre-eclampsia prevention/treatment and rank their potential based on the TPPs. METHODS Adis Insight, Pharmaprojects, WHO international clinical trials registry platform (ICTRP), PubMed and grant databases were searched (Jan-May 2021). The AIM database was screened for all candidates being investigated for pre-eclampsia. Candidates in clinical development were evaluated against nine prespecified criteria from TPPs identified as key for wide-scale implementation, and classified as high, medium or low potential based on matching to the TPPs. Preclinical candidates were categorised by product type, archetype and medicine subclass. RESULTS The AIM database identified 153 candidates for pre-eclampsia. Of the 87 candidates in clinical development, seven were classified as high potential (prevention: esomeprazole, L-arginine, chloroquine, vitamin D and metformin; treatment: sulfasalazine and metformin) and eight as medium potential (prevention: probiotic lactobacilli, dalteparin, selenium and omega-3 fatty acid; treatment: sulforaphane, pravastatin, rosuvastatin and vitamin B3). Sixty-six candidates were in preclinical development, the most common being amino acid/peptides, siRNA-based medicines and polyphenols. CONCLUSIONS This is a novel, evidence-informed approach to identifying promising candidates for pre-eclampsia prevention and treatment - a vital step in stimulating R&D of new medicines for pre-eclampsia suitable for real-world implementation.
Collapse
Affiliation(s)
- Annie R A McDougall
- Maternal, Child and Adolescent Health Program, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia.
| | - Roxanne Hastie
- Department of Obstetrics and Gynaecology, University of Melbourne, Heidelberg, Australia
| | | | | | - Stephen Tong
- Department of Obstetrics and Gynaecology, University of Melbourne, Heidelberg, Australia
| | | | | | - Joshua P Vogel
- Maternal, Child and Adolescent Health Program, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia.,School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| |
Collapse
|
39
|
Louis JM, Parchem J, Vaught A, Tesfalul M, Kendle A, Tsigas E. Preeclampsia: a report and recommendations of the workshop of the Society for Maternal-Fetal Medicine and the Preeclampsia Foundation. Am J Obstet Gynecol 2022; 227:B2-B24. [PMID: 39491898 DOI: 10.1016/j.ajog.2022.06.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Preeclampsia is a substantial cause of perinatal and maternal morbidity and mortality. The prevalence of this condition has increased over the past several decades. Additional opportunities are needed to foster interdisciplinary collaborations and improve patient care in the setting of preeclampsia. In recognition of the Preeclampsia Foundation's 20th anniversary and its work to advance preeclampsia research and clinical agendas, a 2-day virtual workshop on preeclampsia was cosponsored by the Society for Maternal-Fetal Medicine and the Preeclampsia Foundation and held January 25-26, 2021 in conjunction with the 41st annual pregnancy meeting. Leaders with expertise in preeclampsia research, obstetrical care, primary care medicine, cardiology, endocrinology, global health, and patient advocacy gathered to discuss preeclampsia prediction, prevention, management, and long-term impacts. The goals of the workshop were to review the following issues and create consensus concerning research and clinical recommendations: This report, developed collaboratively between the SMFM and the Preeclampsia Foundation, presents the key findings and consensus-based recommendations from the workshop participants.
Collapse
|
40
|
Zhang X, Chen Y, Sun D, Zhu X, Ying X, Yao Y, Fei W, Zheng C. Emerging pharmacologic interventions for pre-eclampsia treatment. Expert Opin Ther Targets 2022; 26:739-759. [PMID: 36223503 DOI: 10.1080/14728222.2022.2134779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Pre-eclampsia is a serious pregnancy complication and a major global concern for the mortality of both mother and fetus. Existing symptomatic treatments do not delay disease progression; thus, timely delivery of the baby is the most effective measure. However, the risk of various maternal and fetal injuries remains. AREAS COVERED In this review, we summarize the potential strategies for pharmacologic interventions in pre-eclamptic therapy. Specifically, we discuss the pathophysiological process of various effective candidate therapeutics that act on potential pathways and molecular targets to inhibit key stages of the disease. We refer to this pathogenesis-focused drug discovery model as a pathogenesis-target-drug (P-T-D) strategy. Finally, we discuss the introduction of nanotechnologies to improve the safety and efficacy of therapeutics via their specific placental targeting ability and placental retention effects. EXPERT OPINION Despite the active development of novel pharmacological treatments based on our current knowledge of pre-eclamptic pathogenesis, investigations are still in the early phase. Thus, further exploration of the pathological mechanisms, integrated with the P-T-D strategy and novel nanosystems, could encourage the development of more effective and safer strategies. Such advances could lead to a shift from expectant management to mechanistic-based therapy for pre-eclampsia.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yue Chen
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Dongli Sun
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiaojun Zhu
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xia Ying
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yao Yao
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| |
Collapse
|
41
|
Dunleavy JEM, Dinh DT, Filby CE, Green E, Hofstee P, Pini T, Rivers N, Skerrett-Byrne DA, Wijayarathna R, Winstanley YE, Zhou W, Richani D. Reproductive biology research down under: highlights from the Australian and New Zealand Annual Meeting of the Society for Reproductive Biology, 2021. Reprod Fertil Dev 2022; 34:855-866. [PMID: 35836362 DOI: 10.1071/rd22115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/17/2022] [Indexed: 11/23/2022] Open
Abstract
Against the backdrop of a global pandemic, the Society for Reproductive Biology (SRB) 2021 meeting reunited the Australian and New Zealand reproductive research community for the first time since 2019 and was the first virtual SRB meeting. Despite the recent global research disruptions, the conference revealed significant advancements in reproductive research, the importance of which span human health, agriculture, and conservation. A core theme was novel technologies, including the use of medical microrobots for therapeutic and sperm delivery, diagnostic hyperspectral imaging, and hydrogel condoms with potential beyond contraception. The importance of challenging the contraceptive status quo was further highlighted with innovations in gene therapies, non-hormonal female contraceptives, epigenetic semen analysis, and in applying evolutionary theory to suppress pest population reproduction. How best to support pregnancies, particularly in the context of global trends of increasing maternal age, was also discussed, with several promising therapies for improved outcomes in assisted reproductive technology, pre-eclampsia, and pre-term birth prevention. The unique insights gained via non-model species was another key focus and presented research emphasised the importance of studying diverse systems to understand fundamental aspects of reproductive biology and evolution. Finally, the meeting highlighted how to effectively translate reproductive research into policy and industry practice.
Collapse
Affiliation(s)
- Jessica E M Dunleavy
- School of BioSciences, Faculty of Science, The University of Melbourne, Parkville, Vic. 3010, Australia
| | - Doan Thao Dinh
- Robinson Research Institute, School of Biomedicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, SA 5006, Australia
| | - Caitlin E Filby
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Vic. 3168, Australia; and Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Vic. 3168, Australia
| | - Ella Green
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, SA 5006, Australia
| | - Pierre Hofstee
- Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Taylor Pini
- School of Veterinary Science, The University of Queensland, Gatton, Qld 4343, Australia
| | - Nicola Rivers
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Vic. 3168, Australia
| | - David A Skerrett-Byrne
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia; and Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton, NSW 2305, Australia
| | - Rukmali Wijayarathna
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Vic. 3168, Australia; and Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, Vic. 3800, Australia
| | - Yasmyn E Winstanley
- Robinson Research Institute, School of Biomedicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, SA 5006, Australia
| | - Wei Zhou
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, Vic. 3010, Australia; and Gynaecology Research Centre, Royal Women's Hospital, Parkville, Vic. 3052, Australia
| | - Dulama Richani
- Fertility & Research Centre, School of Women's and Children's Health, University of New South Wales, Sydney, NSW 2031, Australia
| |
Collapse
|
42
|
Tarry-Adkins JL, Robinson IG, Reynolds RM, Aye ILMH, Charnock-Jones DS, Jenkins B, Koulmann A, Ozanne SE, Aiken CE. Impact of Metformin Treatment on Human Placental Energy Production and Oxidative Stress. Front Cell Dev Biol 2022; 10:935403. [PMID: 35784487 PMCID: PMC9247405 DOI: 10.3389/fcell.2022.935403] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 05/20/2022] [Indexed: 02/02/2023] Open
Abstract
Metformin is increasingly prescribed in pregnancy, with beneficial maternal effects. However, it is not known how metformin-treatment impacts metabolism and energy production in the developing feto-placental unit. We assessed the human placental response to metformin using both in vivo and in vitro treated samples. trophoblasts were derived from placentas collected from non-laboured Caesarean deliveries at term, then treated in vitro with metformin (0.01 mM, 0.1 mM or vehicle). Metformin-concentrations were measured using liquid-chromatography mass-spectrometry. Oxygen consumption in cultured-trophoblasts was measured using a Seahorse-XF Mito Stress Test. Markers of oxidative-stress were assayed using qRT-PCR. Metformin-transporter mRNA and protein-levels were determined by quantitative RT-PCR and Western-blotting respectively. Metformin concentrations were also measured in sample trios (maternal plasma/fetal plasma/placental tissue) from pregnancies exposed to metformin on clinical-grounds. Maternal and fetal metformin concentrations in vivo were highly correlated over a range of concentrations (R2 = 0.76, p < 0.001; average fetal:maternal ratio 1.5; range 0.8-2.1). Basal respiration in trophoblasts was reduced by metformin treatment (0.01 mM metformin; p < 0.05, 0.1 mM metformin; p < 0.001). Mitochondrial-dependent ATP production and proton leak were reduced after treatment with metformin (p < 0.001). Oxidative stress markers were significantly reduced in primary-trophoblast-cultures following treatment with metformin. There is a close linear relationship between placental, fetal, and maternal metformin concentrations. Primary-trophoblast cultures exposed to clinically-relevant metformin concentrations have reduced mitochondrial-respiration, mitochondrial-dependent ATP-production, and reduced markers of oxidative-stress. Given the crucial role of placental energy-production in supporting fetal growth and well-being during pregnancy, the implications of these findings are concerning for intrauterine fetal growth and longer-term metabolic programming in metformin-exposed pregnancies.
Collapse
Affiliation(s)
- Jane L. Tarry-Adkins
- Department of Obstetrics and Gynaecology, The Rosie Hospital and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - India G. Robinson
- Department of Obstetrics and Gynaecology, The Rosie Hospital and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Rebecca M. Reynolds
- Queen’s Medical Research Institute, Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Irving L. M. H. Aye
- Department of Obstetrics and Gynaecology, The Rosie Hospital and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - D. Stephen Charnock-Jones
- Department of Obstetrics and Gynaecology, The Rosie Hospital and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Benjamin Jenkins
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, Addenbrooke’s Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Albert Koulmann
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, Addenbrooke’s Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Susan E. Ozanne
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, Addenbrooke’s Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Catherine E. Aiken
- Department of Obstetrics and Gynaecology, The Rosie Hospital and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, Addenbrooke’s Hospital, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
43
|
Zhang ML, Yang Q, Zhu YD, Zhang YD, Zhang R, Liu J, Zhao XY, Dang QY, Huang DX, Zhang MY, Wei YC, Hu Z, Cai XX, Gao LF, Shan Y, Yu HL. Nobiletin Inhibits Hypoxia-Induced Placental Damage via Modulating P53 Signaling Pathway. Nutrients 2022; 14:nu14112332. [PMID: 35684132 PMCID: PMC9183106 DOI: 10.3390/nu14112332] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/24/2022] [Accepted: 05/30/2022] [Indexed: 11/19/2022] Open
Abstract
In this study, we aimed to evaluate the effect of Nobiletin (NOB) on the placenta of Sprague–Dawley (SD) rats that had undergone reduced uterine perfusion pressure (RUPP) surgery and to evaluate the safety of NOB intervention during pregnancy. The results showed that NOB alleviated placental hypoxia, attenuated placental cell apoptosis, and inhibited placental damage in RUPP rats. No side effect of NOB intervention during pregnancy was observed. BeWo cell lines with P53 knockdown were then constructed using lentiviral transfection, and the P53 signaling pathway was found to be essential for NOB to reduce hypoxia-induced apoptosis of the BeWo cell lines. In summary, NOB attenuated hypoxia-induced placental damage by regulating the P53 signaling pathway, and those findings may contribute some insights into the role of NOB in placental development and the prevention of placental-related diseases.
Collapse
Affiliation(s)
- Meng-Ling Zhang
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (M.-L.Z.); (Q.Y.); (Y.-D.Z.); (Y.-D.Z.); (X.-Y.Z.); (Q.-Y.D.); (D.-X.H.); (M.-Y.Z.); (Y.-C.W.); (Z.H.); (X.-X.C.); (L.-F.G.)
- Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Hunan Provincial Key Laboratory for Fruits and Vegetables Storage Processing and Quality Safety, Changsha 410082, China;
- Hunan Provincial Key Laboratory for Fruits and Vegetables Storage Processing and Quality Safety, Changsha 410082, China
- Hunan Province International Joint Lab on Fruits & Vegetables Processing, Quality and Safety, Changsha 410082, China
- Longping Branch Graduate School, Hunan University, Changsha 410082, China
| | - Qian Yang
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (M.-L.Z.); (Q.Y.); (Y.-D.Z.); (Y.-D.Z.); (X.-Y.Z.); (Q.-Y.D.); (D.-X.H.); (M.-Y.Z.); (Y.-C.W.); (Z.H.); (X.-X.C.); (L.-F.G.)
| | - Yan-Di Zhu
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (M.-L.Z.); (Q.Y.); (Y.-D.Z.); (Y.-D.Z.); (X.-Y.Z.); (Q.-Y.D.); (D.-X.H.); (M.-Y.Z.); (Y.-C.W.); (Z.H.); (X.-X.C.); (L.-F.G.)
| | - Ya-Di Zhang
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (M.-L.Z.); (Q.Y.); (Y.-D.Z.); (Y.-D.Z.); (X.-Y.Z.); (Q.-Y.D.); (D.-X.H.); (M.-Y.Z.); (Y.-C.W.); (Z.H.); (X.-X.C.); (L.-F.G.)
| | - Rui Zhang
- School of Medical Humanity, Peking University, Beijing 100191, China;
| | - Jian Liu
- Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Hunan Provincial Key Laboratory for Fruits and Vegetables Storage Processing and Quality Safety, Changsha 410082, China;
- Hunan Provincial Key Laboratory for Fruits and Vegetables Storage Processing and Quality Safety, Changsha 410082, China
- Hunan Province International Joint Lab on Fruits & Vegetables Processing, Quality and Safety, Changsha 410082, China
- Longping Branch Graduate School, Hunan University, Changsha 410082, China
| | - Xiao-Yan Zhao
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (M.-L.Z.); (Q.Y.); (Y.-D.Z.); (Y.-D.Z.); (X.-Y.Z.); (Q.-Y.D.); (D.-X.H.); (M.-Y.Z.); (Y.-C.W.); (Z.H.); (X.-X.C.); (L.-F.G.)
| | - Qin-Yu Dang
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (M.-L.Z.); (Q.Y.); (Y.-D.Z.); (Y.-D.Z.); (X.-Y.Z.); (Q.-Y.D.); (D.-X.H.); (M.-Y.Z.); (Y.-C.W.); (Z.H.); (X.-X.C.); (L.-F.G.)
| | - Dong-Xu Huang
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (M.-L.Z.); (Q.Y.); (Y.-D.Z.); (Y.-D.Z.); (X.-Y.Z.); (Q.-Y.D.); (D.-X.H.); (M.-Y.Z.); (Y.-C.W.); (Z.H.); (X.-X.C.); (L.-F.G.)
| | - Ming-Yuan Zhang
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (M.-L.Z.); (Q.Y.); (Y.-D.Z.); (Y.-D.Z.); (X.-Y.Z.); (Q.-Y.D.); (D.-X.H.); (M.-Y.Z.); (Y.-C.W.); (Z.H.); (X.-X.C.); (L.-F.G.)
| | - Yu-Chen Wei
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (M.-L.Z.); (Q.Y.); (Y.-D.Z.); (Y.-D.Z.); (X.-Y.Z.); (Q.-Y.D.); (D.-X.H.); (M.-Y.Z.); (Y.-C.W.); (Z.H.); (X.-X.C.); (L.-F.G.)
| | - Zhuo Hu
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (M.-L.Z.); (Q.Y.); (Y.-D.Z.); (Y.-D.Z.); (X.-Y.Z.); (Q.-Y.D.); (D.-X.H.); (M.-Y.Z.); (Y.-C.W.); (Z.H.); (X.-X.C.); (L.-F.G.)
| | - Xia-Xia Cai
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (M.-L.Z.); (Q.Y.); (Y.-D.Z.); (Y.-D.Z.); (X.-Y.Z.); (Q.-Y.D.); (D.-X.H.); (M.-Y.Z.); (Y.-C.W.); (Z.H.); (X.-X.C.); (L.-F.G.)
| | - Li-Fang Gao
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (M.-L.Z.); (Q.Y.); (Y.-D.Z.); (Y.-D.Z.); (X.-Y.Z.); (Q.-Y.D.); (D.-X.H.); (M.-Y.Z.); (Y.-C.W.); (Z.H.); (X.-X.C.); (L.-F.G.)
| | - Yang Shan
- Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Hunan Provincial Key Laboratory for Fruits and Vegetables Storage Processing and Quality Safety, Changsha 410082, China;
- Hunan Provincial Key Laboratory for Fruits and Vegetables Storage Processing and Quality Safety, Changsha 410082, China
- Hunan Province International Joint Lab on Fruits & Vegetables Processing, Quality and Safety, Changsha 410082, China
- Longping Branch Graduate School, Hunan University, Changsha 410082, China
- Correspondence: (Y.S.); (H.-L.Y.); Tel.: +86-731-84691289 (Y.S.); +86-10-83911652 (H.-L.Y.)
| | - Huan-Ling Yu
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (M.-L.Z.); (Q.Y.); (Y.-D.Z.); (Y.-D.Z.); (X.-Y.Z.); (Q.-Y.D.); (D.-X.H.); (M.-Y.Z.); (Y.-C.W.); (Z.H.); (X.-X.C.); (L.-F.G.)
- Correspondence: (Y.S.); (H.-L.Y.); Tel.: +86-731-84691289 (Y.S.); +86-10-83911652 (H.-L.Y.)
| |
Collapse
|
44
|
Diagnostic utility of serial circulating placental growth factor levels and uterine artery Doppler waveforms in diagnosing underlying placental diseases in pregnancies at high risk of placental dysfunction. Am J Obstet Gynecol 2022; 227:618.e1-618.e16. [PMID: 35644246 DOI: 10.1016/j.ajog.2022.05.043] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Placental pathology assessment following delivery in pregnancies complicated by preeclampsia, fetal growth restriction, abruption, and stillbirth reveals a range of underlying diseases. The most common pathology is maternal vascular malperfusion, characterized by high-resistance uterine artery Doppler waveforms and abnormal expression of circulating maternal angiogenic growth factors. Rare placental diseases (massive perivillous fibrinoid deposition and chronic histiocytic intervillositis) are reported to have high recurrence risks, but their associations with uterine artery Doppler waveforms and angiogenic growth factors are presently ill-defined. OBJECTIVE To characterize the patterns of serial placental growth factor measurements and uterine artery Doppler waveform assessments in pregnancies that develop specific types of placental pathology to gain insight into their relationships with the timing of disease onset and pregnancy outcomes. STUDY DESIGN A retrospective cohort study conducted between January 2017 and November 2021 included all singleton pregnancies with at least 1 measurement of maternal circulating placental growth factor between 16 and 36 weeks' gestation, delivery at our institution, and placental pathology analysis demonstrating diagnostic features of maternal vascular malperfusion, fetal vascular malperfusion, villitis of unknown etiology, chronic histiocytic intervillositis, or massive perivillous fibrinoid deposition. Profiles of circulating placental growth factor as gestational age advanced were compared between these placental pathologies. Maternal and perinatal outcomes were recorded. RESULTS A total of 337 pregnancies from 329 individuals met our inclusion criteria. These comprised placental pathology diagnoses of maternal vascular malperfusion (n=109), fetal vascular malperfusion (n=87), villitis of unknown etiology (n=96), chronic histiocytic intervillositis (n=16), and massive perivillous fibrinoid deposition (n=29). Among patients who developed maternal vascular malperfusion, placental growth factor levels gradually declined as pregnancy progressed (placental growth factor <10th percentile at 16-20 weeks' gestation in 42.9%; 20-24 weeks in 61.9%; 24-28 weeks in 77%; and 28-32 weeks in 81.4%) accompanied by mean uterine artery Doppler pulsatility index >95th percentile in 71.6% cases. Patients who developed either fetal vascular malperfusion or villitis of unknown etiology mostly exhibited normal circulating placental growth factor values in association with normal uterine artery Doppler waveforms (mean [standard deviation] pulsatility index values: fetal vascular malperfusion, 1.14 [0.49]; villitis of unknown etiology, 1.13 [0.45]). Patients who developed either chronic histiocytic intervillositis or massive perivillous fibrinoid deposition exhibited persistently low placental growth factor levels from the early second trimester (placental growth factor <10th centile at 16-20 weeks' gestation in 80% and 77.8%, respectively; 20-24 weeks in 88.9% and 63.6%; 24-28 weeks in 85.7% and 75%), all in combination with normal uterine artery Doppler waveforms (mean pulsatility index >95th centile: chronic histiocytic intervillositis, 25%; massive perivillous fibrinoid deposition, 37.9%). Preeclampsia developed in 83 of 337 (24.6%) patients and was most common in those developing maternal vascular malperfusion (54/109, 49.5%) followed by chronic histiocytic intervillositis (7/16, 43.8%). There were 29 stillbirths in the cohort (maternal vascular malperfusion, n=10 [9.2%]; fetal vascular malperfusion, n=5 [5.7%]; villitis of unknown etiology, n=1 [1.0%]; chronic histiocytic intervillositis, n=7 [43.8%]; massive perivillous fibrinoid deposition, n=6 [20.7%]). Most patients experiencing stillbirth exhibited normal uterine artery Doppler waveforms (21/29, 72.4%) and had nonmaternal vascular malperfusion pathologies (19/29, 65.5%). By contrast, 28 of 29 (96.5%) patients experiencing stillbirth had ≥1 low placental growth factor values before fetal death. CONCLUSION Serial circulating maternal placental growth factor tests, in combination with uterine artery Doppler waveform assessments in the second trimester, may indicate the likely underlying type of placental pathology mediating severe adverse perinatal events. This approach has the potential to test disease-specific therapeutic strategies to improve clinical outcomes. Serial placental growth factor testing, compared with uterine artery Doppler studies, identifies a greater proportion of patients destined to have a poor perinatal outcome because diseases other than maternal vascular malperfusion are characterized by normal uteroplacental circulation.
Collapse
|
45
|
Affiliation(s)
- Laura A Magee
- From the Department of Women and Children's Health, School of Life Course Sciences, King's College London (L.A.M., K.H.N., P.D.), the Institute of Women and Children's Health, King's Health Partners Academic Health Science Centre (L.A.M., P.D.), and the Harris Birthright Research Centre for Fetal Medicine, King's College Hospital (K.H.N.) - all in London
| | - Kypros H Nicolaides
- From the Department of Women and Children's Health, School of Life Course Sciences, King's College London (L.A.M., K.H.N., P.D.), the Institute of Women and Children's Health, King's Health Partners Academic Health Science Centre (L.A.M., P.D.), and the Harris Birthright Research Centre for Fetal Medicine, King's College Hospital (K.H.N.) - all in London
| | - Peter von Dadelszen
- From the Department of Women and Children's Health, School of Life Course Sciences, King's College London (L.A.M., K.H.N., P.D.), the Institute of Women and Children's Health, King's Health Partners Academic Health Science Centre (L.A.M., P.D.), and the Harris Birthright Research Centre for Fetal Medicine, King's College Hospital (K.H.N.) - all in London
| |
Collapse
|
46
|
Frühe Präeklampsie: Metformin könnte Geburtstermin hinauszögern. Dtsch Med Wochenschr 2022. [DOI: 10.1055/a-1665-1424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
47
|
Muglia L, Tong S, Ozanne S, Benhalima K. Maternal factors during pregnancy influencing maternal, fetal and childhood outcomes: Meet the Guest Editors. BMC Med 2022; 20:114. [PMID: 35264147 PMCID: PMC8908555 DOI: 10.1186/s12916-022-02294-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 02/09/2022] [Indexed: 01/04/2023] Open
Affiliation(s)
- Louis Muglia
- Burroughs Wellcome Fund, Research Triangle Park, NC, USA.
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Stephen Tong
- Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Perinatal, Mercy Hospital for Women, Melbourne, Australia
| | - Susan Ozanne
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Katrien Benhalima
- Department of Endocrinology, University Hospital Gasthuisberg, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| |
Collapse
|
48
|
Pei J, Liu Z, Wang C, Chu N, Liu L, Tang Y, Liu H, Xiang Q, Cheng H, Li M, Gu W. Progesterone Attenuates SIRT1-Deficiency-Mediated Pre-Eclampsia. Biomolecules 2022; 12:biom12030422. [PMID: 35327614 PMCID: PMC8946184 DOI: 10.3390/biom12030422] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/07/2022] [Accepted: 03/07/2022] [Indexed: 11/16/2022] Open
Abstract
Pre-eclampsia is a severe hypertensive disorder of pregnancy (HDP), mainly characterized by new-onset hypertension with proteinuria after 20-week gestation. Sirtuin1 (SIRT1), a class III histone deacetylase, is associated with the regulation of various pathophysiological processes, including inflammation, immune response, metabolism, and autophagy. However, the effect of SIRT1 in the pathogenesis of pre-eclampsia remains to be elucidated. In this study, we found that the expression of SIRT1 was relatively lower in the placentas and serum samples of pre-eclampsia patients. Typical pre-eclampsia-like symptoms, such as hypertension, proteinuria, fetal growth restriction, kidney injury, and a narrow placental labyrinth layer, were observed in SIRT1 knockdown (SIRT1+/−) mice. Of note, these performances could be improved after the intraperitoneal injection of SIRT1 agonist SRT2104. More importantly, we found that the efficacy of progesterone on attenuating symptoms of PE was profoundly better than that of metformin in SIRT1+/− mice. In addition, our results suggested that progesterone can promote the invasion and inhibit the apoptosis of trophoblasts. These data suggest that SIRT1 plays an important role in pre-eclampsia and that progesterone alleviates pre-eclampsia-like symptoms mediated by SIRT1 deficiency.
Collapse
Affiliation(s)
- Jiangnan Pei
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (J.P.); (Z.L.); (C.W.); (N.C.); (L.L.); (Y.T.); (H.L.)
| | - Zhenzhen Liu
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (J.P.); (Z.L.); (C.W.); (N.C.); (L.L.); (Y.T.); (H.L.)
| | - Chengjie Wang
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (J.P.); (Z.L.); (C.W.); (N.C.); (L.L.); (Y.T.); (H.L.)
| | - Nan Chu
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (J.P.); (Z.L.); (C.W.); (N.C.); (L.L.); (Y.T.); (H.L.)
| | - Lei Liu
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (J.P.); (Z.L.); (C.W.); (N.C.); (L.L.); (Y.T.); (H.L.)
| | - Yao Tang
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (J.P.); (Z.L.); (C.W.); (N.C.); (L.L.); (Y.T.); (H.L.)
| | - Haiyan Liu
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (J.P.); (Z.L.); (C.W.); (N.C.); (L.L.); (Y.T.); (H.L.)
| | - Qianqian Xiang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China;
| | - Haidong Cheng
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (J.P.); (Z.L.); (C.W.); (N.C.); (L.L.); (Y.T.); (H.L.)
- Correspondence: (H.C.); (M.L.); (W.G.)
| | - Mingqing Li
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (J.P.); (Z.L.); (C.W.); (N.C.); (L.L.); (Y.T.); (H.L.)
- Correspondence: (H.C.); (M.L.); (W.G.)
| | - Weirong Gu
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; (J.P.); (Z.L.); (C.W.); (N.C.); (L.L.); (Y.T.); (H.L.)
- Correspondence: (H.C.); (M.L.); (W.G.)
| |
Collapse
|
49
|
Circulating SPINT1 Is Reduced in a Preeclamptic Cohort with Co-Existing Fetal Growth Restriction. J Clin Med 2022; 11:jcm11040901. [PMID: 35207174 PMCID: PMC8877863 DOI: 10.3390/jcm11040901] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/01/2022] [Accepted: 02/06/2022] [Indexed: 02/01/2023] Open
Abstract
Fetal growth restriction (FGR), when undetected antenatally, is the biggest risk factor for preventable stillbirth. Maternal circulating SPINT1 is reduced in pregnancies, which ultimately deliver small for gestational age (SGA) infants at term (birthweight < 10th centile), compared to appropriate for gestational age (AGA) infants (birthweight ≥ 10th centile). SPINT1 is also reduced in FGR diagnosed before 34 weeks’ gestation. We hypothesised that circulating SPINT1 would be decreased in co-existing preterm preeclampsia and FGR. Plasma SPINT1 was measured in samples obtained from two double-blind, randomised therapeutic trials. In the Preeclampsia Intervention with Esomeprazole trial, circulating SPINT1 was decreased in women with preeclampsia who delivered SGA infants (n = 75, median = 18,857 pg/mL, IQR 10,782–29,890 pg/mL, p < 0.0001), relative to those delivering AGA (n = 22, median = 40,168 pg/mL, IQR 22,342–75,172 pg/mL). This was confirmed in the Preeclampsia Intervention 2 with metformin trial where levels of SPINT1 in maternal circulation were reduced in SGA pregnancies (n = 95, median = 57,764 pg/mL, IQR 42,212–91,356 pg/mL, p < 0.0001) compared to AGA controls (n = 40, median = 107,062 pg/mL, IQR 70,183–176,532 pg/mL). Placental Growth Factor (PlGF) and sFlt-1 were also measured. PlGF was significantly reduced in the SGA pregnancies, while ratios of sFlt-1/SPINT1 and sFlt1/PlGF were significantly increased. This is the first study to demonstrate significantly reduced SPINT1 in co-existing FGR and preeclamptic pregnancies.
Collapse
|
50
|
Jung E, Romero R, Yeo L, Gomez-Lopez N, Chaemsaithong P, Jaovisidha A, Gotsch F, Erez O. The etiology of preeclampsia. Am J Obstet Gynecol 2022; 226:S844-S866. [PMID: 35177222 PMCID: PMC8988238 DOI: 10.1016/j.ajog.2021.11.1356] [Citation(s) in RCA: 230] [Impact Index Per Article: 76.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 02/08/2023]
Abstract
Preeclampsia is one of the "great obstetrical syndromes" in which multiple and sometimes overlapping pathologic processes activate a common pathway consisting of endothelial cell activation, intravascular inflammation, and syncytiotrophoblast stress. This article reviews the potential etiologies of preeclampsia. The role of uteroplacental ischemia is well-established on the basis of a solid body of clinical and experimental evidence. A causal role for microorganisms has gained recognition through the realization that periodontal disease and maternal gut dysbiosis are linked to atherosclerosis, thus possibly to a subset of patients with preeclampsia. The recent reports indicating that SARS-CoV-2 infection might be causally linked to preeclampsia are reviewed along with the potential mechanisms involved. Particular etiologic factors, such as the breakdown of maternal-fetal immune tolerance (thought to account for the excess of preeclampsia in primipaternity and egg donation), may operate, in part, through uteroplacental ischemia, whereas other factors such as placental aging may operate largely through syncytiotrophoblast stress. This article also examines the association between gestational diabetes mellitus and maternal obesity with preeclampsia. The role of autoimmunity, fetal diseases, and endocrine disorders is discussed. A greater understanding of the etiologic factors of preeclampsia is essential to improve treatment and prevention.
Collapse
Affiliation(s)
- Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI; Detroit Medical Center, Detroit, MI.
| | - Lami Yeo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI
| | - Piya Chaemsaithong
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Faculty of Medicine, Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Adithep Jaovisidha
- Faculty of Medicine, Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Francesca Gotsch
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Offer Erez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Obstetrics and Gynecology, HaEmek Medical Center, Afula, Israel
| |
Collapse
|