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
|
Coskun E, Ekmekci OB, Gungor Z, Tuten A, Oncul M, Hamzaoğlu K, Gok K, Ekmekci H. Evaluation of vascular peroxidase 1, humanin, MOTS-c and miR-200c expression levels in untreated preeclampsia patients. Mol Biol Rep 2024; 52:66. [PMID: 39704971 DOI: 10.1007/s11033-024-10148-w] [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: 07/16/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND The objective of this study was to evaluate the levels of Vascular Peroxidase 1 (VPO1), humanin, and MOTS-c in relation to miR-200c expression in untreated preeclamptic pregnancies, and to compare these findings with endoglin levels. METHODS AND RESULTS In this study, blood samples were collected from preeclamptic patients presenting to the clinic prior to the initiation of treatment. The levels of endoglin, VPO1, humanin, and MOTS-c were measured using enzyme-linked immunosorbent assay (ELISA), while miR-200c expression was quantified using reverse transcription polymerase chain reaction (RT-PCR). Receiver operating characteristic (ROC) analysis was performed to assess diagnostic accuracy. Statistical significance was determined at p < 0.05. The levels of endoglin, VPO1, and miR-200c were found to be significantly elevated in the preeclampsia group compared to the control group (p < 0.05), whereas MOTS-c levels were significantly reduced (p < 0.05). No significant difference was observed in humanin levels between the two groups. A positive correlation was identified between endoglin levels and VPO1 (r = 0.943, p < 0.001), humanin (r = 0.421, p < 0.01), and uric acid (r = 0.314, p = 0.02) in the preeclamptic group. CONCLUSIONS Our findings suggest that the elevation of VPO1 and miR-200c levels, along with the reduction of humanin and MOTS-c levels, may contribute to the increased endoglin levels and subsequent endothelial dysfunction observed in preeclampsia. These changes may be associated with the pathogenesis and severity of the disease.
Collapse
Affiliation(s)
- Erkam Coskun
- Department of Medical Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey.
| | - Ozlem Balci Ekmekci
- Department of Medical Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Zeynep Gungor
- Department of Medical Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Abdullah Tuten
- Department of Obstetrics and Gynecology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Mahmut Oncul
- Department of Obstetrics and Gynecology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Kubra Hamzaoğlu
- Department of Obstetrics and Gynecology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Koray Gok
- Department of Perinatology, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| | - Hakan Ekmekci
- Department of Medical Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
3
|
Ramos A, Youssef L, Molina P, Torramadé-Moix S, Martinez-Sanchez J, Moreno-Castaño AB, Blasco M, Guillén-Olmos E, De Moner B, Pino M, Tortajada M, Camacho M, Borrell M, Crovetto F, Ramirez-Bajo MJ, Ventura-Aguiar P, Banon-Maneus E, Rovira J, Escolar G, Carreras E, Gratacos E, Diaz-Ricart M, Crispi F, Palomo M. Circulating extracellular vesicles and neutrophil extracellular traps contribute to endothelial dysfunction in preeclampsia. Front Immunol 2024; 15:1488127. [PMID: 39735539 PMCID: PMC11671372 DOI: 10.3389/fimmu.2024.1488127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/28/2024] [Indexed: 12/31/2024] Open
Abstract
Background Preeclampsia (PE) is a pregnancy complication characterized by hypertension, proteinuria, endothelial dysfunction, and complement dysregulation. Placenta-derived extracellular vesicles (EVs), necessary in maternal-fetal communication, might contribute to PE pathogenesis. Moreover, neutrophil extracellular traps (NETs) play a pathogenic role in other complement-mediated pathologies, and their contribution in PE remains unexplored. Materials and methods EVs were isolated from PE (peEVs) and normotensive pregnant women sera. NETs were obtained incubating donor-pre-activated neutrophils with PE or control sera. Microvascular (HMEC) endothelial cells (ECs) were incubated with PE or control sera with or without (depleted sera) EVs or NETs, to assess changes in VCAM-1, ICAM-1, VE-cadherin, eNOS, VWF, ROS, and C5b-9 deposits. Results were expressed as fold increase vs. control. Results VWF, VCAM-1, and ROS expression was significantly higher in cells exposed to PE sera vs. control (12.3 ± 8.1, 3.6 ± 2.3, and 1.8 ± 0.2, respectively, p < 0.05), though significantly lower in cells exposed to depleted PE (dPE) sera (6.1 ± 2.7, 0.7 ± 0.6, and 1.2 ± 0.1, respectively, vs. control, p < 0.05). EC exposure to depleted control sera supplemented with peEVs (dC+peEVs) significantly increased VWF, VCAM-1, and ROS compared to non-supplemented sera (4.5 ± 0.3, 2.8 ± 2.0, and 1.4 ± 0.2, respectively, p < 0.05). ICAM-1, VE-cadherin, and C5b-9 did not differ among groups. ECs incubated with PE-NETs increased VWF and VCAM-1 and decreased VE-cadherin expression vs. control (4 ± 1.6, 5.9 ± 1.2, and 0.5 ± 0.1, respectively, p < 0.05), and notably increased C5b-9 deposit (7.5 ± 2.9, p < 0.05). ICAM-1 and ROS did not differ. Conclusions Both circulating EVs and NETs from PE pregnant women exhibit a deleterious effect on ECs. Whereas EVs trigger a pro-oxidant and proinflammatory state, NETs potentiate the activation of the complement system, as already described in PE.
Collapse
Affiliation(s)
- Alex Ramos
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lina Youssef
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain
| | - Patricia Molina
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Sergi Torramadé-Moix
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Julia Martinez-Sanchez
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ana Belen Moreno-Castaño
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Miquel Blasco
- Nephrology and Kidney Transplant Department, Center of Reference in Complex Glomerular Disease (CSUR), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Red de Investigación Cooperativa Orientada a Resultados en Salud RICORS2040, Universidad Autónoma de Madrid, Madrid, Spain
| | - Elena Guillén-Olmos
- Nephrology and Kidney Transplant Department, Center of Reference in Complex Glomerular Disease (CSUR), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Blanca De Moner
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain
| | - Marc Pino
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Marta Tortajada
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Marta Camacho
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Maria Borrell
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Francesca Crovetto
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Maria Jose Ramirez-Bajo
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Pedro Ventura-Aguiar
- Nephrology and Kidney Transplant Department, Center of Reference in Complex Glomerular Disease (CSUR), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Elisenda Banon-Maneus
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jordi Rovira
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Gines Escolar
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Enric Carreras
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain
| | - Eduard Gratacos
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Instituto de Salud Carlos III, Madrid, Spain
| | - Maribel Diaz-Ricart
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Fatima Crispi
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Palomo
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Hematology External Quality Assessment Laboratory, Biomedical Diagnostic Center, Hospital Clinic of Barcelona, Barcelona, Spain
| |
Collapse
|
4
|
Turan Butun T, Özen N, Ozturk N, Yildirim A, Kilavuz E, Karadag C, Aykan Yuksel B, Basrali F, Karadag B, Ulker P. Red blood cell in preeclampsia: attenuated nitric oxide generation and enhanced reactive oxygen species formation and eryptosis. Scand J Clin Lab Invest 2024; 84:379-390. [PMID: 39321099 DOI: 10.1080/00365513.2024.2394982] [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: 04/25/2024] [Revised: 07/22/2024] [Accepted: 08/18/2024] [Indexed: 09/27/2024]
Abstract
Preeclampsia (PE) pathogenesis is strongly related to diminished nitric oxide (NO) bioavailability and enhanced oxidative stress. Emerging evidence suggests that red blood cells (RBCs) eNOS enzyme contributes to systemic NO bioavailability by its ability of both NO and ROS generation. We aimed to investigate RBC eNOS enzyme activity, NO and ROS generation capacity, eryptosis index and aggregation levels in preeclamptic and uncomplicated pregnant women. Fifty-eight PE patients and 36 healthy pregnant women were included to the investigation. RBC eNOS enzyme activity, intracellular NO, calcium and ROS concentrations and eryptosis levels were determined via flow cytometric methods. RBC deformability and aggregation were measured via LORRCA. Intracellular NO and phosphorylated RBC eNOS levels decreased in PE group compared to healthy pregnant group (p < 0.05, p < 0.001 respectively). Intracellular ROS and calcium levels, eryptosis values and aggregation indexes in the PE group were significantly higher than healthy pregnant group (p < 0.05, p < 0.01, p < 0.05, p < 0.05 respectively). Our results demonstrate for the first time that RBC produce lower NO and higher ROS under PE conditions. Further, RBC of PE patients were more prone to eryptosis and aggregation compared to control group. Our results suggest that, in addition to endothelial cells, RBC also contribute to decreased plasma NO bioavailability via producing less NO and high ROS in PE. Considering increased tendency to eryptosis and aggregation, RBC seem to play role in haemodynamic changes of PE pathogenesis.
Collapse
Affiliation(s)
- Tülay Turan Butun
- Department of Obstetrics and Gynecology, Antalya Training and Research Hospital, Antalya, Türkiye
| | - Nur Özen
- Department of Basic Medical Sciences, Dentistry Faculty, Antalya Bilim University, Antalya, Türkiye
| | - Nihal Ozturk
- Department of Biophysics, Medical Faculty, Akdeniz University, Antalya, Türkiye
| | - Ahmet Yildirim
- Department of Physiology, Medical Faculty, Akdeniz University, Antalya, Türkiye
| | - Ece Kilavuz
- Department of Physiology, Medical Faculty, Akdeniz University, Antalya, Türkiye
| | - Ceyda Karadag
- Department of Obstetrics and Gynecology, Antalya Training and Research Hospital, Antalya, Türkiye
| | - Burcu Aykan Yuksel
- Department of Obstetrics and Gynecology, Antalya Training and Research Hospital, Antalya, Türkiye
| | - Filiz Basrali
- Department of Physiology, Medical Faculty, Akdeniz University, Antalya, Türkiye
| | - Burak Karadag
- Department of Obstetrics and Gynecology, Antalya Training and Research Hospital, Antalya, Türkiye
| | - Pinar Ulker
- Department of Physiology, Medical Faculty, Akdeniz University, Antalya, Türkiye
| |
Collapse
|
5
|
Wu Y, Sun T, Medina P, Narasimhan P, Stevenson DK, Von Versen-Höynck F, Armstrong J, Wu JC, Sayed N, Winn VD. A Novel Stem Cell Model to Study Preeclampsia Endothelial Dysfunction. Reprod Sci 2024; 31:2993-3003. [PMID: 39179924 PMCID: PMC11438721 DOI: 10.1007/s43032-024-01590-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 05/08/2024] [Indexed: 08/26/2024]
Abstract
Preeclampsia is a common pregnancy complication affecting 5% to 7% of all pregnancies worldwide annually. While the pathogenesis is not fully understood, maternal endothelium dysfunction is thought to be a central component to preeclampsia development. Studies to dissect maternal endothelial dysfunction, particularly on a patient-specific basis, are hampered by limited access to systemic primary endothelial cells (ECs). The objective of this study was to establish a replenishable, patient-specific in vitro EC model to allow robust mechanistic studies to dissect endothelial dysfunction in preeclampsia. Induced pluripotent stem cells (iPSCs) from three women with a history of normotensive pregnancies were differentiated into ECs. The established ECs were exposed to pooled sera from normotensive pregnancies, preeclamptic pregnancies, normotensive postpartum for non-pregnant comparison and controls. Endothelial functions including nitric oxide (NO) release, cell migration, tube formation and viability were evaluated. Levels of NO release were significantly lower after incubation with preeclamptic sera compared to the fetal bovine serum (FBS) control, and normotensive and non-pregnant (postpartum) sera treatments were also lower than FBS but higher than preeclamptic sera treatments. Tube formation and cell migration were also impaired with preeclamptic sera compared to FBS controls. Cell viabilities remained unaffected by any sera treatment. Consistent outcomes were obtained across all three patient-specific lines treated with the same pooled sera. Establishment of patient-derived iPSC-ECs treated with pregnancy sera serves as a novel model to explore the interplay between individual maternal endothelial health and circulating factors that lead to endothelial dysfunction in preeclampsia.
Collapse
Affiliation(s)
- Yanming Wu
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Tianyanxin Sun
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Pedro Medina
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Purnima Narasimhan
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - David K Stevenson
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Frauke Von Versen-Höynck
- Department of Obstetrics, Gynecology and Reproductive Sciences, Hannover Medical School, Hannover, Germany
| | - Jennifer Armstrong
- Department of Pediatrics, Section of Neurology and Department of Obstetrics and Gynecology, Division of Basic Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nazish Sayed
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
6
|
Eticha TG, Berhe S, Deressa A, Firdisa D, Tura AK. Determinants of preeclampsia among women who gave birth at Hiwot Fana Comprehensive Specialized University Hospital, Eastern Ethiopia: a case-control study. Sci Rep 2024; 14:18744. [PMID: 39138269 PMCID: PMC11322304 DOI: 10.1038/s41598-024-69622-x] [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: 02/24/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024] Open
Abstract
Pre-eclampsia and eclampsia are the second leading causes of maternal mortality and morbidity. It also results in high perinatal mortality and morbidity. Since eclampsia is preceded by preeclampsia and shows the progression of the disease, they share the same pathogenesis and determining factors. The purpose of this study was to determine determinants of preeclampsia, since it is essential for its prevention and/or its associated consequences. An unmatched case-control study was conducted from September 1-30, 2023 among women who gave birth from June 1, 2020, to August 31, 2023, at Hiwot Fana Comprehensive Specialized University Hospital. Women who had preeclampsia were considered cases, while those without were controls. The sample size was calculated using EPI Info version 7 for a case-control study using the following assumptions: 95% confidence interval, power of 80%, case-to-control ratio of 1:2, and 5% non-response rate were 305. Data was collected using Google Form, and analyzed using SPSS version 26. Variables that had a p-value of < 0.05 on multivariable logistic regression were considered statistically significant, and their association was explained using an odds ratio at a 95% confidence interval. A total of 300 women (100 cases and 200 controls) with a mean age of 24.4 years were included in the study. Rural residence (AOR 2.04, 95% CI 1.10-3.76), age less than 20 years (AOR 3.04, 95% CI 1.58-5.85), history of hypertensive disorders of pregnancy (AOR 5.52, 95% CI 1.76-17.33), and no antenatal care (AOR 2.38, 95% CI 1.19-4.75) were found to be the determinants of preeclampsia. We found that living in a rural areas, previous history of preeclampsia, no antenatal care, and < 20 years of age were significantly associated with preeclampsia. In addition to previous preeclampsia, younger and rural resident pregnant women should be given attention in preeclampsia screening and prevention.
Collapse
Affiliation(s)
- Tadesse Gure Eticha
- Department of Obstetrics and Gynecology, School of Medicine, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia.
| | - Solomon Berhe
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Alemayehu Deressa
- School of Public Health, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Dawit Firdisa
- School of Public Health, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Abera Kenay Tura
- School of Nursing and Midwifery, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
- Department of International Health, Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
7
|
Lu C, Zheng F, Pan L, Han Q, Wu J, Zhang W. Suppression of circular RNA serum and glucocorticoid-induced kinase 1 elevates antioxidant molecules and angiogenesis in trophoblast cells to attenuate preeclampsia via microRNA-508-3p to target and restrain PUM homolog 1. J Obstet Gynaecol Res 2024; 50:322-333. [PMID: 38081797 DOI: 10.1111/jog.15843] [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: 02/09/2023] [Accepted: 11/20/2023] [Indexed: 03/04/2024]
Abstract
AIM Preeclampsia (PE) is a pregnancy-specific syndrome characterized by hypertension and proteinuria. Recently, multiple circular RNAs (circRNAs) were considered latent clinical diagnostic markers or therapeutic targets. This study was to explore the impact of circRNA serum and glucocorticoid-induced kinase 1 (SGK1) on PE via influencing the microRNA (miR)-508-3p/PUM homolog 1 (PUM1) axis. METHODS Placental tissues of 34 pregnant women with PE and 34 normal pregnant women were collected to measure circRNA SGK1 (circSGK1), miR-508-3p, and PUM1. Human placental trophoblasts HTR-8/SVneo were transfected with plasmids, thereafter to observe proliferation, migration, invasion, and apoptosis, analyze antioxidant molecules Troxerutin (TXN), Glutamate-cysteine ligase catalytic subunit (GCLC), NAD (P) H-quinone oxidoreductase 1 (NQO1), and determine angiogenesis. After the construction of the PE rat model, antioxidant molecules TXN, GCLC, and NQO1, vascular-associated factor vascular endothelial growth factor A (VEGF-A), and histopathological conditions were tested. Examination of the binding of circSGK1 and miR-508-3p with PUM1 was performed. RESULTS Our data showed that circSGK1 expression was elevated in the placenta of patients with PE. Silenced circSGK1 or elevated miR-508-3p promoted the growth and antioxidant molecules and angiogenesis in trophoblast cells; CircSGK1 combined with miR-508-3p, and miR-508-3p targeted PUM1. CONCLUSIONS In summary, suppression of circSGK1 augments antioxidant molecules and angiogenesis in trophoblast cells to attenuate PE via miR-508-3p to target PUM1.
Collapse
Affiliation(s)
- CaiXia Lu
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - FuMin Zheng
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - LiXiao Pan
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - QingFang Han
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - JiXia Wu
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - WenKe Zhang
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
8
|
Yagel S, Cohen SM, Admati I, Skarbianskis N, Solt I, Zeisel A, Beharier O, Goldman-Wohl D. Expert review: preeclampsia Type I and Type II. Am J Obstet Gynecol MFM 2023; 5:101203. [PMID: 37871693 DOI: 10.1016/j.ajogmf.2023.101203] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/25/2023]
Abstract
Pregnancy involves an interplay between maternal and fetal factors affecting changes to maternal anatomy and physiology to support the developing fetus and ensure the well-being of both the mother and offspring. A century of research has provided evidence of the imperative role of the placenta in the development of preeclampsia. Recently, a growing body of evidence has supported the adaptations of the maternal cardiovascular system during normal pregnancy and its maladaptation in preeclampsia. Debate surrounds the roles of the placenta vs the maternal cardiovascular system in the pathophysiology of preeclampsia. We proposed an integrated model of the maternal cardiac-placental-fetal array and the development of preeclampsia, which reconciles the disease phenotypes and their proposed origins, whether placenta-dominant or maternal cardiovascular system-dominant. These phenotypes are sufficiently diverse to define 2 distinct types: preeclampsia Type I and Type II. Type I preeclampsia may present earlier, characterized by placental dysfunction or malperfusion, shallow trophoblast invasion, inadequate spiral artery conversion, profound syncytiotrophoblast stress, elevated soluble fms-like tyrosine kinase-1 levels, reduced placental growth factor levels, high peripheral vascular resistance, and low cardiac output. Type I is more often accompanied by fetal growth restriction, and low placental growth factor levels have a measurable impact on maternal cardiac remodeling and function. Type II preeclampsia typically occurs in the later stages of pregnancy and entails an evolving maternal cardiovascular intolerance to the demands of pregnancy, with a moderately dysfunctional placenta and inadequate blood supply. The soluble fms-like tyrosine kinase-1-placental growth factor ratio may be normal or slightly disturbed, peripheral vascular resistance is low, and cardiac output is high, but these adaptations still fail to meet demand. Emergent placental dysfunction, coupled with an increasing inability to meet demand, more often appears with fetal macrosomia, multiple pregnancies, or prolonged pregnancy. Support for the notion of 2 types of preeclampsia observable on the molecular level is provided by single-cell transcriptomic survey of gene expression patterns across different cell classes. This revealed widespread dysregulation of gene expression across all cell types, and significant imbalance in fms-like tyrosine kinase-1 (FLT1) and placental growth factor, particularly marked in the syncytium of early preeclampsia cases. Classification of preeclampsia into Type I and Type II can inform future research to develop targeted screening, prevention, and treatment approaches.
Collapse
Affiliation(s)
- Simcha Yagel
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Hebrew University Faculty of Medicine, Jerusalem, Israel (Dr Yagel, Ms Cohen, and Drs Beharier and Goldman-Wohl).
| | - Sarah M Cohen
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Hebrew University Faculty of Medicine, Jerusalem, Israel (Dr Yagel, Ms Cohen, and Drs Beharier and Goldman-Wohl)
| | - Inbal Admati
- Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, Israel (Ms Admati, Mr Skarbianskis, and Dr Zeisel)
| | - Niv Skarbianskis
- Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, Israel (Ms Admati, Mr Skarbianskis, and Dr Zeisel)
| | - Ido Solt
- Department of Obstetrics and Gynecology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel (Dr Solt)
| | - Amit Zeisel
- Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, Israel (Ms Admati, Mr Skarbianskis, and Dr Zeisel)
| | - Ofer Beharier
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Hebrew University Faculty of Medicine, Jerusalem, Israel (Dr Yagel, Ms Cohen, and Drs Beharier and Goldman-Wohl)
| | - Debra Goldman-Wohl
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Hebrew University Faculty of Medicine, Jerusalem, Israel (Dr Yagel, Ms Cohen, and Drs Beharier and Goldman-Wohl)
| |
Collapse
|
9
|
Ricci CA, Reid DM, Sun J, Santillan DA, Santillan MK, Phillips NR, Goulopoulou S. Maternal and fetal mitochondrial gene dysregulation in hypertensive disorders of pregnancy. Physiol Genomics 2023; 55:275-285. [PMID: 37184228 PMCID: PMC10292966 DOI: 10.1152/physiolgenomics.00005.2023] [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: 01/29/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 05/16/2023] Open
Abstract
Mitochondrial dysfunction has been implicated in pregnancy-induced hypertension (PIH). The role of mitochondrial gene dysregulation in PIH, and consequences for maternal-fetal interactions, remain elusive. Here, we investigated mitochondrial gene expression and dysregulation in maternal and placental tissues from pregnancies with and without PIH; further, we measured circulating mitochondrial DNA (mtDNA) mutational load, an index of mtDNA integrity. Differential gene expression analysis followed by Time Course Gene Set Analysis (TcGSA) was conducted on publicly available high throughput sequencing transcriptomic data sets. Mutational load analysis was carried out on peripheral mononuclear blood cells from healthy pregnant individuals and individuals with preeclampsia. Thirty mitochondrial differentially expressed genes (mtDEGs) were detected in the maternal cell-free circulating transcriptome, whereas nine were detected in placental transcriptome from pregnancies with PIH. In PIH pregnancies, maternal mitochondrial dysregulation was associated with pathways involved in inflammation, cell death/survival, and placental development, whereas fetal mitochondrial dysregulation was associated with increased production of extracellular vesicles (EVs) at term. Mothers with preeclampsia did not exhibit a significantly different degree of mtDNA mutational load. Our findings support the involvement of maternal mitochondrial dysregulation in the pathophysiology of PIH and suggest that mitochondria may mediate maternal-fetal interactions during healthy pregnancy.NEW & NOTEWORTHY This study identifies aberrant maternal and fetal expression of mitochondrial genes in pregnancies with gestational hypertension and preeclampsia. Mitochondrial gene dysregulation may be a common etiological factor contributing to the development of de novo hypertension in pregnancy-associated hypertensive disorders.
Collapse
Affiliation(s)
- Contessa A Ricci
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Danielle M Reid
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Jie Sun
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Donna A Santillan
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
| | - Mark K Santillan
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
| | - Nicole R Phillips
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Styliani Goulopoulou
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
- Department of Gynecology and Obstetrics, Lawrence D. Longo MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California, United States
| |
Collapse
|
10
|
Yagel S, Cohen SM, Goldman-Wohl D, Beharier O. Redefining pre-eclampsia as Type I or II: implementing an integrated model of the maternal-cardiovascular-placental-fetal array. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2023; 61:293-301. [PMID: 36378064 DOI: 10.1002/uog.26121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/04/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Affiliation(s)
- S Yagel
- Division of Obstetrics and Gynecology, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - S M Cohen
- Division of Obstetrics and Gynecology, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - D Goldman-Wohl
- Division of Obstetrics and Gynecology, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - O Beharier
- Division of Obstetrics and Gynecology, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
11
|
Mukherjee I, Singh S, Karmakar A, Kashyap N, Mridha AR, Sharma JB, Luthra K, Sharma RS, Biswas S, Dhar R, Karmakar S. New immune horizons in therapeutics and diagnostic approaches to Preeclampsia. Am J Reprod Immunol 2023; 89:e13670. [PMID: 36565013 DOI: 10.1111/aji.13670] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 11/02/2022] [Accepted: 12/10/2022] [Indexed: 12/25/2022] Open
Abstract
Hypertensive disorders of pregnancy (HDP) are one of the commonest maladies, affecting 5%-10% of pregnancies worldwide. The American College of Obstetricians and Gynecologists (ACOG) identifies four categories of HDP, namely gestational hypertension (GH), Preeclampsia (PE), chronic hypertension (CH), and CH with superimposed PE. PE is a multisystem, heterogeneous disorder that encompasses 2%-8% of all pregnancy-related complications, contributing to about 9% to 26% of maternal deaths in low-income countries and 16% in high-income countries. These translate to 50 000 maternal deaths and over 500 000 fetal deaths worldwide, therefore demanding high priority in understanding clinical presentation, screening, diagnostic criteria, and effective management. PE is accompanied by uteroplacental insufficiency leading to vascular and metabolic changes, vasoconstriction, and end-organ ischemia. PE is diagnosed after 20 weeks of pregnancy in women who were previously normotensive or hypertensive. Besides shallow trophoblast invasion and inadequate remodeling of uterine arteries, dysregulation of the nonimmune system has been the focal point in PE. This results from aberrant immune system activation and imbalanced differentiation of T cells. Further, a failure of tolerance toward the semi-allogenic fetus results due to altered distribution of Tregs such as CD4+FoxP3+ or CD4+CD25+CD127(low) FoxP3+ cells, thereby creating a cytotoxic environment by suboptimal production of immunosuppressive cytokines like IL-10, IL-4, and IL-13. Also, intracellular production of complement protein C5a may result in decreased FoxP3+ regulatory T cells. With immune system dysfunction as a major driver in PE pathogenesis, it is logical that therapeutic targeting of components of the immune system with pharmacologic agents like anti-inflammatory and immune-modulating molecules are either being used or under clinical trial. Cholesterol synthesis inhibitors like Pravastatin may improve placental perfusion in PE, while Eculizumab (monoclonal antibody inhibiting C5) and small molecular inhibitor of C5a, Zilucoplan are under investigation. Monoclonal antibody against IL-17(Secukinumab) has been proposed to alter the Th imbalance in PE. Autologous Treg therapy and immune checkpoint inhibitors like anti-CTLA-4 are emerging as new candidates in immune horizons for PE management in the future.
Collapse
Affiliation(s)
- Indrani Mukherjee
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.,Amity Institute of Biotechnology (AIB), Amity University, Noida, India
| | - Sunil Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Abhibrato Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Neha Kashyap
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Asit Ranjan Mridha
- Department of Obstetrics & Gynaecology, All India Institute of Medical Sciences, New Delhi, India
| | - Jai Bhagwan Sharma
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Radhey Shyam Sharma
- Ex-Head and Scientist G, Indian Council of Medical Research, New Delhi, India
| | - Subhrajit Biswas
- Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University, Noida, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
12
|
De Bie FR, Basurto D, Kumar S, Deprest J, Russo FM. Sildenafil during the 2nd and 3rd Trimester of Pregnancy: Trials and Tribulations. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:11207. [PMID: 36141480 PMCID: PMC9517616 DOI: 10.3390/ijerph191811207] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 06/16/2023]
Abstract
Sildenafil, a phosphodiesterase 5 inhibitor with a vasodilatory and anti-remodeling effect, has been investigated concerning various conditions during pregnancy. Per indication, we herein review the rationale and the most relevant experimental and clinical studies, including systematic reviews and meta-analyses, when available. Indications for using sildenafil during the second and third trimester of pregnancy include maternal pulmonary hypertension, preeclampsia, preterm labor, fetal growth restriction, oligohydramnios, fetal distress, and congenital diaphragmatic hernia. For most indications, the rationale for administering prenatal sildenafil is based on limited, equivocal data from in vitro studies and rodent disease models. Clinical studies report mild maternal side effects and suggest good fetal tolerance and safety depending on the underlying pathology.
Collapse
Affiliation(s)
| | - David Basurto
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Sailesh Kumar
- Mater Research Institute and School of Medicine, University of Queensland, Brisbane, QLD 4343, Australia
| | - Jan Deprest
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Department of Obstetrics and Gynecology, UZ Leuven, 3000 Leuven, Belgium
| | - Francesca Maria Russo
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Department of Obstetrics and Gynecology, UZ Leuven, 3000 Leuven, Belgium
| |
Collapse
|
13
|
Perusquía M. Androgens and Non-Genomic vascular responses in hypertension. Biochem Pharmacol 2022; 203:115200. [PMID: 35926652 DOI: 10.1016/j.bcp.2022.115200] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/02/2022]
Abstract
Arterial hypertension is a global public health concern. In the last few years, the interest in androgen deficiency has been growing, and the association between androgens and high blood pressure (BP) is still controversial. One purpose of this review was to summarize the available findings in order to clarify whether male sex steroid hormones have beneficial or harmful effect on BP. The second purpose was to enhance the recognition of the acute non-genomic sex-independent vasorelaxing effect of androgens. Remarkably, BP variation is expected to be a consequence of the androgen-induced vasorelaxation which reduces systemic BP; hence the in vivo vasodepressor, hypotensive, and antihypertensive responses of androgens were also analyzed. This article reviews the current understanding of the physiological regulation of vascular smooth muscle contractility by androgens. Additionally, it summarizes older and more recent data on androgens, and some of the possible underlying mechanisms of relaxation, structural-functional differences in the androgen molecules, and their designing ability to induce vasorelaxation. The clinical relevance of these findings in terms of designing future therapeutics mainly the 5-reduced metabolite of testosterone, 5β-dihydrotestosterone, is also highlighted. Literature collected through a PubMed database search, as well as our experimental work, was used for the present review.
Collapse
Affiliation(s)
- Mercedes Perusquía
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510 Ciudad de México, México.
| |
Collapse
|
14
|
Narang K, Weaver AL, Mehta RA, Garovic VD, Szymanski LM. Intraabdominal pressure as a marker for physiologic and pathologic processes in pregnancy. Hypertens Pregnancy 2022; 41:181-189. [DOI: 10.1080/10641955.2022.2085741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Kavita Narang
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Amy L. Weaver
- Division of Clinical Trial and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Ramila A. Mehta
- Division of Clinical Trial and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Vesna D. Garovic
- Division of Nephrology and Hypertension, Department of Internal Medicine, and Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Linda M. Szymanski
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
15
|
Sethuraman V, Pu Y, Gingrich J, Jing J, Long R, Olomu IN, Veiga-Lopez A. Expression of ABC transporters during syncytialization in preeclampsia. Pregnancy Hypertens 2022; 27:181-188. [PMID: 35124425 PMCID: PMC9017055 DOI: 10.1016/j.preghy.2022.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/29/2021] [Accepted: 01/23/2022] [Indexed: 01/12/2023]
Abstract
Preeclampsia complicates 2-8% of pregnancies and is associated with prematurity and intrauterine growth restriction. Cholesterol and sterol transport is a key function of the placenta and it is elicited through ATP binding cassette (ABC) transporters. ABCA1 expression changes during trophoblast cell fusion, a process required to form the placental syncytium that enables maternal-fetal nutrient transfer. ABCA1 expression is dysregulated in preeclamptic placentas. But whether ABC transporters expression during trophoblast fusion is disrupted in preeclampsia remains unknown. We investigated if cholesterol and sterol ABC transporters are altered in term and preterm preeclampsia placentas and during human cytotrophoblast syncytialization. Human placental biopsies were collected from healthy term (≥37 weeks; n = 11) and term preeclamptic (≥36 6/7 weeks; n = 8) and pre-term preeclamptic (28-35 weeks; n = 8) pregnancies. Both, protein and mRNA expression for ABCA1, ABCG1, ABCG5, and ABCG8 were evaluated. Primary cytotrophoblasts isolated from a subset of placentas were induced to syncytialize for 96 h and ABCA1, ABCG1 and ABCG8 mRNA expression evaluated at 0 h and 96 h. Protein and gene expression of ABC transporters were not altered in preeclamptic placentas. In the healthy Term group, ABCA1 expression was similar before and after syncytialization. After 96 h of syncytialization, mRNA expression of ABCA1 and ABCG1 increased significantly, while ABCG8 decreased significantly in term-preeclampsia, but not pre-term preeclampsia. While placental expression of ABCA1 and ABCG1 remained unaltered in term preeclampsia, the disruption in their dynamic expression pattern during cytotrophoblast syncytialization suggests that cholesterol transport may contribute to the pathophysiologic role of the placenta in preeclampsia.
Collapse
Affiliation(s)
- Visalakshi Sethuraman
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Yong Pu
- Department of Pathology, University of Illinois at Chicago
| | - Jeremy Gingrich
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, Michigan, USA
| | - Jiongjie Jing
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, Michigan, USA
| | - Robert Long
- Department of Obstetrics and Gynecology, Sparrow Health System, East Lansing, Michigan, USA
| | - Isoken Nicholas Olomu
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Almudena Veiga-Lopez
- Department of Pathology, University of Illinois at Chicago, USA; Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
16
|
Khaing A, Swe AT, Aung CL, Thwin MM, Sein MT. Expression of Endothelin-1 and Endothelial Nitric Oxide Synthase in Normal and Preeclamptic Placentae. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRICIA : REVISTA DA FEDERACAO BRASILEIRA DAS SOCIEDADES DE GINECOLOGIA E OBSTETRICIA 2022; 44:125-132. [PMID: 35213910 PMCID: PMC9948136 DOI: 10.1055/s-0042-1742317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVE To investigate the expression of endothelin-1 (ET-1) and endothelial nitric oxide (NO) synthase (eNOS) in normal and preeclamptic (PE) placentae. METHODS The present cross-sectional analytical study was performed in normal and PE primigravidae (n = 10 in each group) who were admitted to the North Okkalapa General and Teaching Hospital from February 2019 to February 2020. Serum samples were collected immediately before delivery, and placental tissues were collected immediately after emergency or elective cesarean section. The expression of placental eNOS was measured by western blot, and the levels of ET-1 in placental tissue homogenates and in the serum were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS The PE group had significantly higher serum levels of ET-1 (median: 116.56 pg/mL; IQR: 89.14-159.62 pg/mL) than the normal group (median: 60.02 pg/mL; IQR: 50.89-94.37 pg/mL) (p < 0.05). However, statistically significant differences were not observed in the levels of ET-1 in placental tissue homogenates between normal and PE placentae (median: 0.007 pg/µg of total protein; IQR: 0.002-0.0123 pg/µg of total protein; and median: 0.005 pg/µg of total protein; IQR: 0.003-0.016 pg/µg of total protein respectively). The median and IQR values of relative placental eNOS expression were significantly higher in the PE group than in the normal group (p < 0.05). The serum levels of ET-1 level were not significantly correlated with placental ET-1 expression, and neither there was a significant correlation between placental ET-1 and eNOS expression in any of the groups. CONCLUSION The serum levels of ET-1 were significantly higher in PE pregnant women compared with normal pregnant women, while the ET-1 levels of placental tissue homogenates were not significantly different. Serum ET-1 rather than placental ET-1 might play a major role in the pathogenesis of PE.
Collapse
Affiliation(s)
- Aung Khaing
- Department of Physiology, University of Medicine 2, Yangon, The Republic of the Union of Myanmar
| | - Aye Thet Swe
- Department of Physiology, University of Medicine 2, Yangon, The Republic of the Union of Myanmar
| | - Cho Lwin Aung
- Department of Physiology, University of Medicine 2, Yangon, The Republic of the Union of Myanmar
| | - Mya Mya Thwin
- Physiology Unit, Universiti Sultan Zanial Abidin, Kuala Terengganu,Terengganu, Malaysia
| | - Mya Thanda Sein
- Department of Physiology, University of Medicine 2, Yangon, The Republic of the Union of Myanmar
| |
Collapse
|
17
|
Gan B, Wu X, Lu L, Li X, Li J. The Value of Prenatal First Systolic Blood Pressure Can Predict Severe Preeclampsia and Birth Weight in Patients With Preeclampsia. Front Med (Lausanne) 2022; 8:771738. [PMID: 35186968 PMCID: PMC8850716 DOI: 10.3389/fmed.2021.771738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/17/2021] [Indexed: 11/16/2022] Open
Abstract
Background Preeclampsia is a serious complication of pregnancy that threatens the safety of the fetus and mother. We assessed the relationship between systolic blood pressure (SBP) in the early pregnancy stage (12 weeks) in patients with preeclampsia and the development of severe eclampsia and birth weight. Methods Patients were categorized based on the quartiles of the prenatal first SBP level. Logistic regression analysis was performed to assess whether prenatal first SBP was a risk factor for low birth weight and severe preeclampsia. The area under the receiver-operating characteristic curve (AUC) of sensitivity and specificity were used to predict the risk of low birth weight and severe preeclampsia. Results A total of 333 patients with preeclampsia were enrolled. There were 162 (48.6%) patients with severe preeclampsia and 270 (81.08%) cesareans. Group I patients with a prenatal first SBP ≤ 119 mmHg prenatal had a higher birth weight. Multiple logistic regression analysis showed that serum creatinine (p = 0.025), prenatal first SBP (p = 0.029), S-preeclampsia (p = 0.003), gestational age (p < 0.001), total cholesterol (TC) (p < 0.001), and low-density lipoprotein (LDL) (p < 0.001) were independent risk factors for low birth weight. Multiple logistic regression analysis showed that prenatal first SBP (p = 0.003), TC (p = 0.002), and B-type natriuretic peptide (BNP) (p < 0.001) were independent risk factors for severe preeclampsia. Compared with Group I (SBP ≤ 119 mmHg), the incidence of low birth weight for patients in groups III (131 ≤ SBP ≤ 138 mmHg) and IV (SBP ≥ 139 mmHg) was significantly higher. Even after correcting for age, gestational age, and biochemical indices, the difference remained statistically significant. The risk of diagnosed severe preeclampsia for patients in Groups IV (SBP ≥ 139 mmHg), III (131 ≤ SBP ≤ 138 mmHg), and II (120 ≤ SBP ≤ 130 mmHg) was significantly higher than that in Group I (SBP ≤ 119 mmHg). The AUC of the prenatal first SBP for predicting low birth weight and severe preeclampsia was 0.676 (95% CI 0.618–0.733, p < 0.001) and 0.727 (95% CI 0.673–0.781, p < 0.001), respectively, in patients with preeclampsia. Conclusions Prenatal first SBP was associated with birth weight and severe preeclampsia. Higher prenatal first SBP in patients with preeclampsia can predict low birth weight and severe preeclampsia.
Collapse
|
18
|
Bodke VV, Burdette JE. Advancements in Microfluidic Systems for the Study of Female Reproductive Biology. Endocrinology 2021; 162:6225875. [PMID: 33852726 PMCID: PMC8571709 DOI: 10.1210/endocr/bqab078] [Citation(s) in RCA: 10] [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: 12/31/2020] [Indexed: 12/11/2022]
Abstract
The female reproductive tract is a highly complex physiological system that consists of the ovaries, fallopian tubes, uterus, cervix, and vagina. An enhanced understanding of the molecular, cellular, and genetic mechanisms of the tract will allow for the development of more effective assisted reproductive technologies, therapeutics, and screening strategies for female specific disorders. Traditional 2-dimensional and 3-dimensional static culture systems may not always reflect the cellular and physical contexts or physicochemical microenvironment necessary to understand the dynamic exchange that is crucial for the functioning of the reproductive system. Microfluidic systems present a unique opportunity to study the female reproductive tract, as these systems recapitulate the multicellular architecture, contacts between different tissues, and microenvironmental cues that largely influence cell structure, function, behavior, and growth. This review discusses examples, challenges, and benefits of using microfluidic systems to model ovaries, fallopian tubes, endometrium, and placenta. Additionally, this review also briefly discusses the use of these systems in studying the effects of endocrine disrupting chemicals and diseases such as ovarian cancer, preeclampsia, and polycystic ovarian syndrome.
Collapse
Affiliation(s)
- Vedant V Bodke
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago 60607, USA
| | - Joanna E Burdette
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago 60607, USA
- Correspondence: Joanna E. Burdette, PhD, University of Illinois at Chicago, 900 S. Ashland Ave, Chicago, IL 60607, USA.
| |
Collapse
|
19
|
Helgadóttir H, Tropea T, Gizurarson S, Mandalà M. Endothelium-Derived Hyperpolarizing Factor (EDHF) Mediates Acetylsalicylic Acid (Aspirin) Vasodilation of Pregnant Rat Mesenteric Arteries. Int J Mol Sci 2021; 22:ijms221810162. [PMID: 34576324 PMCID: PMC8471567 DOI: 10.3390/ijms221810162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 01/12/2023] Open
Abstract
Acetylsalicylic acid (aspirin) exhibits a broad range of activities, including analgesic, antipyretic, and antiplatelet properties. Recent clinical studies also recommend aspirin prophylaxis in women with a high risk of pre-eclampsia, a major complication of pregnancy characterized by hypertension. We investigated the effect of aspirin on mesenteric resistance arteries and found outdiscovered the molecular mechanism underlying this action. Aspirin (10−12–10−6 M) was tested on pregnant rat mesenteric resistance arteries by a pressurized arteriography. Aspirin was investigated in the presence of several inhibitors of: (a) nitric oxide synthase (L-NAME 2 × 10−4 M); (b) cyclooxygenase (Indomethacin, 10−5 M); (c) Ca2+-activated K+ channels (Kca): small conductance (SKca, Apamin, 10−7 M), intermediate conductance (IKca, TRAM34, 10−5 M), and big conductance (BKca, paxilline, 10−5 M); and (d) endothelial-derived hyperpolarizing factor (high KCl, 80 mM). Aspirin caused a concentration-dependent vasodilation. Aspirin-vasodilation was abolished by removal of endothelium or by high KCl. Furthermore, preincubation with either apamin plus TRAM-34 or paxillin significantly attenuated aspirin vasodilation (p < 0.05). For the first time, we showed that aspirin induced endothelium-dependent vasodilation in mesenteric resistance arteries through the endothelial-derived hyperpolarizing factor (EDHF) and calcium-activated potassium channels. By activating this molecular mechanism, aspirin may lower peripheral vascular resistance and be beneficial in pregnancies complicated by hypertension.
Collapse
Affiliation(s)
- Helga Helgadóttir
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy; (H.H.); (T.T.)
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, 107 Reykjavik, Iceland;
| | - Teresa Tropea
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy; (H.H.); (T.T.)
| | - Sveinbjörn Gizurarson
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, 107 Reykjavik, Iceland;
| | - Maurizio Mandalà
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy; (H.H.); (T.T.)
- Correspondence:
| |
Collapse
|
20
|
Hiersch L, Ray JG, Barrett J, Berger H, Geary M, McDonald SD, Diong C, Gandhi S, Guan J, Murray-Davis B, Melamed N. Maternal cardiovascular disease after twin pregnancies complicated by hypertensive disorders of pregnancy: a population-based cohort study. CMAJ 2021; 193:E1448-E1458. [PMID: 34544783 PMCID: PMC8476218 DOI: 10.1503/cmaj.202837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2021] [Indexed: 12/15/2022] Open
Abstract
Background: People whose singleton pregnancy is affected by hypertensive disorders of pregnancy (HDP) are at risk of future cardiovascular disease. It is unclear, however, whether this association can be extrapolated to twin pregnancies. We aimed to compare the association between HDP and future cardiovascular disease after twin and singleton pregnancies. Methods: We conducted a population-based retrospective cohort study that included nulliparous people in Ontario, Canada, 1992–2017. We compared the future risk of cardiovascular disease among pregnant people from the following 4 groups: those who delivered a singleton without HDP (referent) and with HDP, and those who delivered twins either with or without HDP. Results: The populations of the 4 groups were as follows: 1 431 651 pregnant people in the singleton birth without HDP group; 98 631 singleton birth with HDP; 21 046 twin birth without HDP; and 4283 twin birth with HDP. The median duration of follow-up was 13 (interquartile range 7–20) years. The incidence rate of cardiovascular disease was lowest among those with a singleton or twin birth without HDP (0.72 and 0.74 per 1000 person-years, respectively). Compared with people with a singleton birth without HDP, the risk of cardiovascular disease was highest among those with a singleton birth and HDP (1.47 per 1000 person-years; adjusted hazard ratio [HR] 1.81 [95% confidence interval (CI) 1.72–1.90]), followed by people with a twin pregnancy and HDP (1.07 per 1000 person-years; adjusted HR 1.36 [95% CI 1.04–1.77]). The risk of the primary outcome after a twin pregnancy with HDP was lower than that after a singleton pregnancy with HDP (adjusted HR 0.74 [95% CI 0.57–0.97]), when compared directly. Interpretation: In a twin pregnancy, HDP are weaker risk factors for postpartum cardiovascular disease than in a singleton pregnancy.
Collapse
Affiliation(s)
- Liran Hiersch
- Division of Maternal-Fetal Medicine (Hiersch, Barrett, Melamed), Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ont.; Departments of Obstetrics and Gynaecology (Hiersch), Lis Maternity Hospital and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Departments of Medicine and Obstetrics and Gynaecology (Ray), St. Michael's Hospital, University of Toronto; ICES Central (Ray, Diong, Gandhi, Guan); Division of Maternal-Fetal Medicine (Berger, Geary), Department of Obstetrics and Gynecology, St. Michael's Hospital, University of Toronto, Toronto, Ont.; Division of Maternal-Fetal Medicine (McDonald, Murray-Davis), Departments of Obstetrics and Gynecology, Radiology, and Research Methods, Evidence & Impact, McMaster University, Hamilton, Ont.
| | - Joel G Ray
- Division of Maternal-Fetal Medicine (Hiersch, Barrett, Melamed), Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ont.; Departments of Obstetrics and Gynaecology (Hiersch), Lis Maternity Hospital and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Departments of Medicine and Obstetrics and Gynaecology (Ray), St. Michael's Hospital, University of Toronto; ICES Central (Ray, Diong, Gandhi, Guan); Division of Maternal-Fetal Medicine (Berger, Geary), Department of Obstetrics and Gynecology, St. Michael's Hospital, University of Toronto, Toronto, Ont.; Division of Maternal-Fetal Medicine (McDonald, Murray-Davis), Departments of Obstetrics and Gynecology, Radiology, and Research Methods, Evidence & Impact, McMaster University, Hamilton, Ont
| | - Jon Barrett
- Division of Maternal-Fetal Medicine (Hiersch, Barrett, Melamed), Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ont.; Departments of Obstetrics and Gynaecology (Hiersch), Lis Maternity Hospital and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Departments of Medicine and Obstetrics and Gynaecology (Ray), St. Michael's Hospital, University of Toronto; ICES Central (Ray, Diong, Gandhi, Guan); Division of Maternal-Fetal Medicine (Berger, Geary), Department of Obstetrics and Gynecology, St. Michael's Hospital, University of Toronto, Toronto, Ont.; Division of Maternal-Fetal Medicine (McDonald, Murray-Davis), Departments of Obstetrics and Gynecology, Radiology, and Research Methods, Evidence & Impact, McMaster University, Hamilton, Ont
| | - Howard Berger
- Division of Maternal-Fetal Medicine (Hiersch, Barrett, Melamed), Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ont.; Departments of Obstetrics and Gynaecology (Hiersch), Lis Maternity Hospital and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Departments of Medicine and Obstetrics and Gynaecology (Ray), St. Michael's Hospital, University of Toronto; ICES Central (Ray, Diong, Gandhi, Guan); Division of Maternal-Fetal Medicine (Berger, Geary), Department of Obstetrics and Gynecology, St. Michael's Hospital, University of Toronto, Toronto, Ont.; Division of Maternal-Fetal Medicine (McDonald, Murray-Davis), Departments of Obstetrics and Gynecology, Radiology, and Research Methods, Evidence & Impact, McMaster University, Hamilton, Ont
| | - Michael Geary
- Division of Maternal-Fetal Medicine (Hiersch, Barrett, Melamed), Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ont.; Departments of Obstetrics and Gynaecology (Hiersch), Lis Maternity Hospital and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Departments of Medicine and Obstetrics and Gynaecology (Ray), St. Michael's Hospital, University of Toronto; ICES Central (Ray, Diong, Gandhi, Guan); Division of Maternal-Fetal Medicine (Berger, Geary), Department of Obstetrics and Gynecology, St. Michael's Hospital, University of Toronto, Toronto, Ont.; Division of Maternal-Fetal Medicine (McDonald, Murray-Davis), Departments of Obstetrics and Gynecology, Radiology, and Research Methods, Evidence & Impact, McMaster University, Hamilton, Ont
| | - Sarah D McDonald
- Division of Maternal-Fetal Medicine (Hiersch, Barrett, Melamed), Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ont.; Departments of Obstetrics and Gynaecology (Hiersch), Lis Maternity Hospital and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Departments of Medicine and Obstetrics and Gynaecology (Ray), St. Michael's Hospital, University of Toronto; ICES Central (Ray, Diong, Gandhi, Guan); Division of Maternal-Fetal Medicine (Berger, Geary), Department of Obstetrics and Gynecology, St. Michael's Hospital, University of Toronto, Toronto, Ont.; Division of Maternal-Fetal Medicine (McDonald, Murray-Davis), Departments of Obstetrics and Gynecology, Radiology, and Research Methods, Evidence & Impact, McMaster University, Hamilton, Ont
| | - Christina Diong
- Division of Maternal-Fetal Medicine (Hiersch, Barrett, Melamed), Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ont.; Departments of Obstetrics and Gynaecology (Hiersch), Lis Maternity Hospital and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Departments of Medicine and Obstetrics and Gynaecology (Ray), St. Michael's Hospital, University of Toronto; ICES Central (Ray, Diong, Gandhi, Guan); Division of Maternal-Fetal Medicine (Berger, Geary), Department of Obstetrics and Gynecology, St. Michael's Hospital, University of Toronto, Toronto, Ont.; Division of Maternal-Fetal Medicine (McDonald, Murray-Davis), Departments of Obstetrics and Gynecology, Radiology, and Research Methods, Evidence & Impact, McMaster University, Hamilton, Ont
| | - Sima Gandhi
- Division of Maternal-Fetal Medicine (Hiersch, Barrett, Melamed), Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ont.; Departments of Obstetrics and Gynaecology (Hiersch), Lis Maternity Hospital and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Departments of Medicine and Obstetrics and Gynaecology (Ray), St. Michael's Hospital, University of Toronto; ICES Central (Ray, Diong, Gandhi, Guan); Division of Maternal-Fetal Medicine (Berger, Geary), Department of Obstetrics and Gynecology, St. Michael's Hospital, University of Toronto, Toronto, Ont.; Division of Maternal-Fetal Medicine (McDonald, Murray-Davis), Departments of Obstetrics and Gynecology, Radiology, and Research Methods, Evidence & Impact, McMaster University, Hamilton, Ont
| | - Jun Guan
- Division of Maternal-Fetal Medicine (Hiersch, Barrett, Melamed), Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ont.; Departments of Obstetrics and Gynaecology (Hiersch), Lis Maternity Hospital and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Departments of Medicine and Obstetrics and Gynaecology (Ray), St. Michael's Hospital, University of Toronto; ICES Central (Ray, Diong, Gandhi, Guan); Division of Maternal-Fetal Medicine (Berger, Geary), Department of Obstetrics and Gynecology, St. Michael's Hospital, University of Toronto, Toronto, Ont.; Division of Maternal-Fetal Medicine (McDonald, Murray-Davis), Departments of Obstetrics and Gynecology, Radiology, and Research Methods, Evidence & Impact, McMaster University, Hamilton, Ont
| | - Beth Murray-Davis
- Division of Maternal-Fetal Medicine (Hiersch, Barrett, Melamed), Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ont.; Departments of Obstetrics and Gynaecology (Hiersch), Lis Maternity Hospital and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Departments of Medicine and Obstetrics and Gynaecology (Ray), St. Michael's Hospital, University of Toronto; ICES Central (Ray, Diong, Gandhi, Guan); Division of Maternal-Fetal Medicine (Berger, Geary), Department of Obstetrics and Gynecology, St. Michael's Hospital, University of Toronto, Toronto, Ont.; Division of Maternal-Fetal Medicine (McDonald, Murray-Davis), Departments of Obstetrics and Gynecology, Radiology, and Research Methods, Evidence & Impact, McMaster University, Hamilton, Ont
| | - Nir Melamed
- Division of Maternal-Fetal Medicine (Hiersch, Barrett, Melamed), Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ont.; Departments of Obstetrics and Gynaecology (Hiersch), Lis Maternity Hospital and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Departments of Medicine and Obstetrics and Gynaecology (Ray), St. Michael's Hospital, University of Toronto; ICES Central (Ray, Diong, Gandhi, Guan); Division of Maternal-Fetal Medicine (Berger, Geary), Department of Obstetrics and Gynecology, St. Michael's Hospital, University of Toronto, Toronto, Ont.; Division of Maternal-Fetal Medicine (McDonald, Murray-Davis), Departments of Obstetrics and Gynecology, Radiology, and Research Methods, Evidence & Impact, McMaster University, Hamilton, Ont
| | | |
Collapse
|
21
|
Effect of Endogenic and Exogenic Oxidative Stress Triggers on Adverse Pregnancy Outcomes: Preeclampsia, Fetal Growth Restriction, Gestational Diabetes Mellitus and Preterm Birth. Int J Mol Sci 2021; 22:ijms221810122. [PMID: 34576285 PMCID: PMC8468091 DOI: 10.3390/ijms221810122] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/10/2021] [Accepted: 09/16/2021] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress is caused by an imbalance between the production of reactive oxygen species (ROS) in cells and tissues and the ability of a biological system to detoxify them. During a normal pregnancy, oxidative stress increases the normal systemic inflammatory response and is usually well-controlled by the balanced body mechanism of the detoxification of anti-oxidative products. However, pregnancy is also a condition in which this adaptation and balance can be easily disrupted. Excessive ROS is detrimental and associated with many pregnancy complications, such as preeclampsia (PE), fetal growth restriction (FGR), gestational diabetes mellitus (GDM), and preterm birth (PTB), by damaging placentation. The placenta is a tissue rich in mitochondria that produces the majority of ROS, so it is important to maintain normal placental function and properly develop its vascular network to ensure a safe and healthy pregnancy. Antioxidants may ameliorate these diseases, and related research is progressing. This review aimed to determine the association between oxidative stress and adverse pregnancy outcomes, especially PE, FGR, GDM, and PTB, and explore how to overcome this oxidative stress in these unfavorable conditions.
Collapse
|
22
|
Chu H, Sacharidou A, Nguyen A, Li C, Chambliss KL, Salmon JE, Shen YM, Lo J, Leone GW, Herz J, Hui DY, Marciano DK, Abrahams VM, Natale BV, Montalbano AP, Xiao X, Xu L, Natale DR, Shaul PW, Mineo C. Protein Phosphatase 2A Activation Via ApoER2 in Trophoblasts Drives Preeclampsia in a Mouse Model of the Antiphospholipid Syndrome. Circ Res 2021; 129:735-750. [PMID: 34404233 DOI: 10.1161/circresaha.120.318941] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Haiyan Chu
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Anastasia Sacharidou
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - An Nguyen
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Chun Li
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Ken L Chambliss
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Jane E Salmon
- Medicine, Hospital for Special Surgery, Weill Cornell Medical College, New York (J.E.S.)
| | - Yu-Min Shen
- Internal Medicine (Y.-M.S., D.K.M.), University of Texas Southwestern Medical Center, Dallas
| | - Julie Lo
- Obstetrics and Gynecology (J.L.), University of Texas Southwestern Medical Center, Dallas
| | - Gustavo W Leone
- Froedtert-Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee (G.W.L.)
| | - Joachim Herz
- Molecular Genetics (J.H.), University of Texas Southwestern Medical Center, Dallas
| | - David Y Hui
- Pathology, University of Cincinnati College of Medicine (D.Y.H.)
| | - Denise K Marciano
- Internal Medicine (Y.-M.S., D.K.M.), University of Texas Southwestern Medical Center, Dallas.,Cell Biology (D.K.M., C.M.), University of Texas Southwestern Medical Center, Dallas
| | - Vikki M Abrahams
- Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT (V.M.A.)
| | - Bryony V Natale
- Obstetrics, Gynecology & Reproductive Science, University of California San Diego, La Jolla (B.V.N., D.R.N.).,Obstetrics and Gynaecology, School of Medicine, Queen's University, Ontario, Canada (B.V.N., D.R.N.)
| | - Alina P Montalbano
- Biochemistry and Obstetrics and Gynecology (A.P.M.), University of Texas Southwestern Medical Center, Dallas
| | - Xue Xiao
- Population and Data Sciences and Pediatrics (X.X., L.X.), University of Texas Southwestern Medical Center, Dallas
| | - Lin Xu
- Population and Data Sciences and Pediatrics (X.X., L.X.), University of Texas Southwestern Medical Center, Dallas
| | - David R Natale
- Obstetrics, Gynecology & Reproductive Science, University of California San Diego, La Jolla (B.V.N., D.R.N.).,Obstetrics and Gynaecology, School of Medicine, Queen's University, Ontario, Canada (B.V.N., D.R.N.)
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.)
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Pediatrics (H.C., A.S., A.N., C.L., K.L.C., P.W.S., C.M.).,Cell Biology (D.K.M., C.M.), University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
23
|
Yeh H, Chiang CC, Yen TH. Hepatocellular carcinoma in patients with renal dysfunction: Pathophysiology, prognosis, and treatment challenges. World J Gastroenterol 2021; 27:4104-4142. [PMID: 34326614 PMCID: PMC8311541 DOI: 10.3748/wjg.v27.i26.4104] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/17/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
The population of patients with hepatocellular carcinoma (HCC) overlaps to a high degree with those for chronic kidney disease (CKD) and end-stage renal disease (ESRD). The degrees of renal dysfunction vary, from the various stages of CKD to dialysis-dependent ESRD, which often affects the prognosis and treatment choice of patients with HCC. In addition, renal dysfunction makes treatment more difficult and may negatively affect treatment outcomes. This study summarized the possible causes of the high comorbidity of HCC and renal dysfunction. The possible mechanisms of CKD causing HCC involve uremia itself, long-term dialysis status, immunosuppressive agents for postrenal transplant status, and miscellaneous factors such as hormone alterations and dysbiosis. The possible mechanisms of HCC affecting renal function include direct tumor invasion and hepatorenal syndrome. Finally, we categorized the risk factors that could lead to both HCC and CKD into four categories: Environmental toxins, viral hepatitis, metabolic syndrome, and vasoactive factors. Both CKD and ESRD have been reported to negatively affect HCC prognosis, but more research is warranted to confirm this. Furthermore, ESRD status itself ought not to prevent patients receiving aggressive treatments. This study then adopted the well-known Barcelona Clinic Liver Cancer guidelines as a framework to discuss the indicators for each stage of HCC treatment, treatment-related adverse renal effects, and concerns that are specific to patients with pre-existing renal dysfunction when undergoing aggressive treatments against CKD and ESRD. Such aggressive treatments include liver resection, simultaneous liver kidney transplantation, radiofrequency ablation, and transarterial chemoembolization. Finally, focusing on patients unable to receive active treatment, this study compiled information on the latest systemic pharmacological therapies, including targeted and immunotherapeutic drugs. Based on available clinical studies and Food and Drug Administration labels, this study details the drug indications, side effects, and dose adjustments for patients with renal dysfunction. It also provides a comprehensive review of information on HCC patients with renal dysfunction from disease onset to treatment.
Collapse
Affiliation(s)
- Hsuan Yeh
- Department of Nephrology, Chang Gung Memorial Hospital and Chang Gung University, Taipei 105, Taiwan
| | - Chun-Cheng Chiang
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Tzung-Hai Yen
- Department of Nephrology, Chang Gung Memorial Hospital and Chang Gung University, Taipei 105, Taiwan
| |
Collapse
|
24
|
Liang Y, Wang F, Chen G, Lu W, Zhang Y. Vasohibin 1, a clinically relevant biomarker, contributes to pre-eclampsia. Int J Clin Pract 2021; 75:e14017. [PMID: 33491841 DOI: 10.1111/ijcp.14017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 01/07/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Pre-eclampsia is a leading health threat for pregnant women which is characterised by hypertension and proteinuria. The detailed mechanism is elusive and no effective therapy is available. Predictive biomarkers are needed for accurate diagnosis. Vasohibin-1 (VASH1) is an intrinsic inhibitor of angiogenesis induced by angiogenic factors in endothelial cells. This study aimed to evaluate the role of VASH1 as a useful biomarker for pre-eclampsia. METHODS VASH1 level was examined by ELISA and immunoblotting assay in the serum and placental samples from healthy pregnant women and pre-eclampsia patients. Cellular assay was performed to assess cell migration and invasion with different levels of VASH1. The level of VASH1 was measured under different oxygen conditions by qPCR. RESULTS VASH1 was highly expressed in the serum and placenta of pre-eclampsia patients. Overexpression of VASH1 led to attenuated cell migration and invasion ability and reduced levels of matrix metallopeptidase 2 and 9. VASH1 was significantly induced in primary human trophoblast cells and placental explants under hypoxic condition in hypoxia-inducible factor 1 alpha-dependent manner. CONCLUSION Our study suggested that VASH1 could be used as a potentially novel biomarker for pre-eclampsia and its level may positively correlate with the disease status.
Collapse
Affiliation(s)
- Yulin Liang
- Department of Obstetrics and gynecology, Heze Municipal Hospital of Shandong Province, Heze, China
| | - Fang Wang
- Department of Obstetrics and gynecology, Heze Municipal Hospital of Shandong Province, Heze, China
| | - Guoru Chen
- Department of Obstetrics and gynecology, Heze Municipal Hospital of Shandong Province, Heze, China
| | - Wei Lu
- Department of Obstetrics and gynecology, Heze Municipal Hospital of Shandong Province, Heze, China
| | - Yonghua Zhang
- Department of Obstetrics and gynecology, Heze Municipal Hospital of Shandong Province, Heze, China
| |
Collapse
|
25
|
Matsuyama T, Tomimatsu T, Mimura K, Yagi K, Kawanishi Y, Kakigano A, Nakamura H, Endo M, Kimura T. Complement activation by an angiogenic imbalance leads to systemic vascular endothelial dysfunction: A new proposal for the pathophysiology of preeclampsia. J Reprod Immunol 2021; 145:103322. [PMID: 33887508 DOI: 10.1016/j.jri.2021.103322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/18/2021] [Accepted: 04/13/2021] [Indexed: 10/21/2022]
Abstract
The underlying mechanism of preeclampsia by which an angiogenic imbalance results in systemic vascular endothelial dysfunction remains unclear. Complement activation directly induces endothelial dysfunction and is known to be involved in preeclampsia; nevertheless, the association between complement activation and angiogenic imbalance has not been established. This study aimed to evaluate whether angiogenic imbalance affects the expression and secretion of inhibitory complement factor H (CFH) in endothelial cells, resulting in complement activation and systemic vascular endothelial dysfunction. Viability of human umbilical vein endothelial cells (HUVECs) was assessed upon CFH knockdown by targeted-siRNA, and were incubated with complement factors. HUVECs were also treated with placental growth factor (PlGF) and/or soluble fms-like tyrosine kinase 1 (sFlt1), and CFH expression and secretion were measured. These cells were evaluated by cell viability assay and cell surface complement activation was quantified by immunocytochemical assessment of C5b-9 deposition. HUVECs transfected with CFH-siRNA had significantly lower viability than that of control cells. Moreover, the expression and secretion of CFH were significantly increased upon PlGF treatment compared with PlGF + sFlt1 combo. HUVECs treated with PlGF had less C5b-9 deposition and higher viability than HUVECs treated with PlGF + sFlt1. In summary, CFH was found to be essential for endothelial cell survival by inhibiting complement activation. An angiogenic imbalance, including decreased PlGF and increased sFlt1, suppresses CFH expression and secretion, resulting in complement activation on the surface of endothelial cells and systemic vascular endothelial dysfunction.
Collapse
Affiliation(s)
- Tatsuya Matsuyama
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takuji Tomimatsu
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| | - Kazuya Mimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kazunobu Yagi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoko Kawanishi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Aiko Kakigano
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hitomi Nakamura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masayuki Endo
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
26
|
Lafuente-Ganuza P, Lequerica-Fernandez P, Carretero F, Escudero AI, Martinez-Morillo E, Sabria E, Herraiz I, Galindo A, Lopez A, Martinez-Triguero ML, Alvarez FV. A more accurate prediction to rule in and rule out pre-eclampsia using the sFlt-1/PlGF ratio and NT-proBNP as biomarkers. Clin Chem Lab Med 2021; 58:399-407. [PMID: 31734648 DOI: 10.1515/cclm-2019-0939] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 10/12/2019] [Indexed: 12/16/2022]
Abstract
Background The management of potential pre-eclamptic patients using the soluble FMS-like tyrosine kinase 1 (sFlt-1)/ placental growth factor (PlGF) ratio is characterised by frequent false-positive results. Methods A retrospective cohort study was conducted to identify and validate cut-off values, obtained using a machine learning model, for the sFlt-1/PlGF ratio and NT-proBNP that would be predictive of the absence or presence of early-onset pre-eclampsia (PE) in singleton pregnancies presenting at 24 to 33 + 6 weeks of gestation. Results For the development cohort, we defined two sFlt-1/PlGF ratio cut-off values of 23 and 45 to rule out and rule in early-onset PE at any time between 24 and 33 + 6 weeks of gestation. Using an sFlt-1/PlGF ratio cut-off value of 23, the negative predictive value (NPV) for the development of early-onset PE was 100% (95% confidence interval [CI]: 99.5-100). The positive predictive value (PPV) of an sFlt-1/PlGF ratio >45 for a diagnosis of early-onset PE was 49.5% (95% CI: 45.8-55.6). When an NT-proBNP value >174 was combined with an sFlt-1/PlGF ratio >45, the PPV was 86% (95% CI: 79.2-92.6). In the validation cohort, the negative and positive values were very similar to those found for the development cohort. Conclusions An sFlt-1/PlGF ratio <23 rules out early-onset PE between 24 and 33 + 6 weeks of gestation at any time, with an NPV of 100%. An sFlt-1/PlGF ratio >45 with an NT-proBNP value >174 significantly enhances the probability of developing early-onset PE.
Collapse
Affiliation(s)
- Paula Lafuente-Ganuza
- Clinical Biochemistry, Laboratory Medicine, Hospital Universitario Central de Asturias, Avenida de Roma, s/n, 33011 Oviedo, Spain
| | - Paloma Lequerica-Fernandez
- Clinical Biochemistry, Laboratory Medicine, Hospital Universitario Central de Asturias, Avenida de Roma, s/n, 33011 Oviedo, Spain
| | | | - Ana I Escudero
- Obstetrics and Gynaecology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Eduardo Martinez-Morillo
- Clinical Biochemistry, Laboratory Medicine, Hospital Universitario Central de Asturias, Avenida de Roma, s/n, 33011 Oviedo, Spain
| | - Enric Sabria
- Obstetrics and Gynaecology Department, Hospital-Residencia Sant Camil, Barcelona, Spain
| | - Ignacio Herraiz
- Fetal Medicine Unit, Maternal and Child Health and Development Network, Department of Obstetrics and Gynaecology, University Hospital 12 de Octubre, Complutense University of Madrid, Madrid, Spain
| | - Alberto Galindo
- Fetal Medicine Unit, Maternal and Child Health and Development Network, Department of Obstetrics and Gynaecology, University Hospital 12 de Octubre, Complutense University of Madrid, Madrid, Spain
| | - Ana Lopez
- Clinical Laboratory, University Hospital 12 de Octubre, Madrid, Spain
| | | | - Francisco V Alvarez
- Clinical Biochemistry, Laboratory Medicine, Hospital Universitario Central de Asturias, Avenida de Roma, s/n, 33011 Oviedo, Spain
- Department of Biochemistry and Molecular Biology, Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
27
|
Todros T, Paulesu L, Cardaropoli S, Rolfo A, Masturzo B, Ermini L, Romagnoli R, Ietta F. Role of the Macrophage Migration Inhibitory Factor in the Pathophysiology of Pre-Eclampsia. Int J Mol Sci 2021; 22:1823. [PMID: 33673075 PMCID: PMC7917653 DOI: 10.3390/ijms22041823] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
Proinflammatory cytokines are produced in pregnancy in response to the invading pathogens and/or nonmicrobial causes such as damage-associated molecules and embryonic semi-allogenic antigens. While inflammation is essential for a successful pregnancy, an excessive inflammatory response is implicated in several pathologies including pre-eclampsia (PE). This review focuses on the proinflammatory cytokine macrophage migration inhibitory factor (MIF), a critical regulator of the innate immune response and a major player of processes allowing normal placental development. PE is a severe pregnancy-related syndrome characterized by exaggerated inflammatory response and generalized endothelial damage. In some cases, usually of early onset, it originates from a maldevelopment of the placenta, and is associated with intrauterine growth restriction (IUGR) (placental PE). In other cases, usually of late onset, pre-pregnancy maternal diseases represent risk factors for the development of the disease (maternal PE). Available data suggest that low MIF production in early pregnancy could contribute to the abnormal placentation. The resulting placental hypoxia in later pregnancy could produce high release of MIF in maternal serum typical of placental PE. More studies are needed to understand the role of MIF, if any, in maternal PE.
Collapse
Affiliation(s)
- Tullia Todros
- Department of Surgical Sciences, University of Turin, Via Ventimiglia 3, 10126 Turin, Italy; (T.T.); (A.R.)
| | - Luana Paulesu
- Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.E.); (R.R.); (F.I.)
| | - Simona Cardaropoli
- Department of Public Health and Pediatrics, University of Turin, 10126 Turin, Italy;
| | - Alessandro Rolfo
- Department of Surgical Sciences, University of Turin, Via Ventimiglia 3, 10126 Turin, Italy; (T.T.); (A.R.)
| | | | - Leonardo Ermini
- Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.E.); (R.R.); (F.I.)
| | - Roberta Romagnoli
- Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.E.); (R.R.); (F.I.)
| | - Francesca Ietta
- Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.E.); (R.R.); (F.I.)
| |
Collapse
|
28
|
Hernandez I, Chissey A, Guibourdenche J, Atasoy R, Coumoul X, Fournier T, Beaudeux JL, Zerrad-Saadi A. Human Placental NADPH Oxidase Mediates sFlt-1 and PlGF Secretion in Early Pregnancy: Exploration of the TGF-β1/p38 MAPK Pathways. Antioxidants (Basel) 2021; 10:antiox10020281. [PMID: 33673360 PMCID: PMC7918586 DOI: 10.3390/antiox10020281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
Preeclampsia, a hypertensive disorder occurring during pregnancy, is characterized by excessive oxidative stress and trophoblast dysfunction with dysregulation of soluble Fms-like tyrosine kinase 1 (sFlt-1) and placental growth factor (PlGF) production. Nicotinamide Adenine Dinucleotide Phosphate (NADPH) oxidase (Nox) is the major source of placental superoxide in early pregnancy and its activation with the subsequent formation of superoxide has been demonstrated for various agents including Transforming Growth Factor beta-1 (TGF-β1), a well-known p38 MAPK pathway activator. However, the bridge between Nox and sFlt-1 remains unknown. The purpose of this study was to explore the possible signaling pathway of TGF-β1/Nox/p38 induced sFlt-1 production in human chorionic villi (CV). Methods: Human chorionic villi from first trimester placenta (7–9 Gestational Weeks (GW)) were treated with TGF-β1 or preincubated with p38 inhibitor, SB203580. For NADPH oxidase inhibition, CV were treated with diphenyleneiodonium (DPI). The protein levels of phospho-p38, p38, phospho-Mothers Against Decapentaplegic homolog 2 (SMAD2), and SMAD2 were detected by Western blot. The secretion of sFlt-1 and PlGF by chorionic villi were measured with Electrochemiluminescence Immunologic Assays, and NADPH oxidase activity was monitored by lucigenin method. Results: We demonstrate for the first time that NADPH oxidase is involved in sFlt-1 and PlGF secretion in first trimester chorionic villi. Indeed, the inhibition of Nox by DPI decreases sFlt-1, and increases PlGF secretions. We also demonstrate the involvement of p38 MAPK in sFlt-1 secretion and Nox activation as blocking the p38 MAPK phosphorylation decreases both sFlt-1 secretion and superoxide production. Nevertheless, TGF-β1-mediated p38 activation do not seem to be involved in regulation of the first trimester placental angiogenic balance and no crosstalk was found between SMAD2 and p38 MAPK pathways. Conclusions: Thus, the placental NADPH oxidase play a major role in mediating the signal transduction cascade of sFlt-1 production. Furthermore, we highlight for the first time the involvement of p38 activation in first trimester placental Nox activity.
Collapse
Affiliation(s)
- Isabelle Hernandez
- Université de Paris, INSERM UMR-S 1139, 3PHM, F-75006 Paris, France; (A.C.); (J.G.); (R.A.); (T.F.); (J.-L.B.)
- Correspondence: (I.H.); (A.Z.-S.); Tel.: +33-1-53-73-96-03 (A.Z.-S.)
| | - Audrey Chissey
- Université de Paris, INSERM UMR-S 1139, 3PHM, F-75006 Paris, France; (A.C.); (J.G.); (R.A.); (T.F.); (J.-L.B.)
| | - Jean Guibourdenche
- Université de Paris, INSERM UMR-S 1139, 3PHM, F-75006 Paris, France; (A.C.); (J.G.); (R.A.); (T.F.); (J.-L.B.)
- UF d’hormonologie Adulte de Cochin AP-HP, Hôpitaux Universitaires, F-75006 Paris, France
| | - Roger Atasoy
- Université de Paris, INSERM UMR-S 1139, 3PHM, F-75006 Paris, France; (A.C.); (J.G.); (R.A.); (T.F.); (J.-L.B.)
| | - Xavier Coumoul
- Université de Paris, INSERM UMR-S 1124, F-75006 Paris, France;
| | - Thierry Fournier
- Université de Paris, INSERM UMR-S 1139, 3PHM, F-75006 Paris, France; (A.C.); (J.G.); (R.A.); (T.F.); (J.-L.B.)
| | - Jean-Louis Beaudeux
- Université de Paris, INSERM UMR-S 1139, 3PHM, F-75006 Paris, France; (A.C.); (J.G.); (R.A.); (T.F.); (J.-L.B.)
| | - Amal Zerrad-Saadi
- Université de Paris, INSERM UMR-S 1139, 3PHM, F-75006 Paris, France; (A.C.); (J.G.); (R.A.); (T.F.); (J.-L.B.)
- Correspondence: (I.H.); (A.Z.-S.); Tel.: +33-1-53-73-96-03 (A.Z.-S.)
| |
Collapse
|
29
|
Tossetta G, Fantone S, Giannubilo SR, Marzioni D. The Multifaced Actions of Curcumin in Pregnancy Outcome. Antioxidants (Basel) 2021; 10:antiox10010126. [PMID: 33477354 PMCID: PMC7830020 DOI: 10.3390/antiox10010126] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/11/2022] Open
Abstract
Curcumin, also known as diferuloylmethane, is the main polyphenolic substance present in the rhizomes of Curcuma longa L. This plant showed many beneficial effects and has been used since ancient times for both food and pharmaceutical purposes. Due to its pleiotropic functions, curcumin consumption in the human diet has become very common thanks also to the fact that this natural compound is considered quite safe as it does not have serious side effects. Its functions as an anti-inflammatory, anti-oxidant, neuroprotective, immunomodulatory, anti-toxicant, anti-apoptotic, and anti-diabetic compound are already known and widely demonstrated. There are numerous studies concerning its effects on various human pathologies including cancer, diabetes and arthritis while the studies on curcumin during pregnancy have been performed only in animal models. Data concerning the role of curcumin as anti-inflammatory compound suggest a possible use of curcumin in managing pregnancy complications such as Preeclampsia (PE), Gestational Diabetes Mellitus (GDM), Fetal Growth Restriction (FGR), PreTerm Birth (PTB), and exposure to toxic agents and pathogens. The aim of this review is to present data to support the possible use of curcumin in clinical trials on human gestation complications.
Collapse
Affiliation(s)
- Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy; (G.T.); (S.F.)
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, 60126 Ancona, Italy;
| | - Sonia Fantone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy; (G.T.); (S.F.)
| | - Stefano Raffaele Giannubilo
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, 60126 Ancona, Italy;
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy; (G.T.); (S.F.)
- Correspondence: ; Tel.:+39-071.2206268
| |
Collapse
|
30
|
Sá CPND, Jiménez MF, Rosa MW, Arlindo EM, Ayub ACK, Cardoso RB, Kreitchmann R, El Beitune P. Evaluation of Angiogenic Factors (PlGF and sFlt-1) in Pre-eclampsia Diagnosis. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRÍCIA 2020; 42:697-704. [PMID: 33254263 PMCID: PMC10309227 DOI: 10.1055/s-0040-1713916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
OBJECTIVE Recent observations support the hypothesis that an imbalance between angiogenic factors has a fundamental role in the pathogenesis of pre-eclampsia and is responsible for the clinical manifestations of the disease. The goal of the present study was to evaluate the sensitivity, specificity, and the best accuracy level of Soluble fms-like tyrosine kinase-1 (sFlt-1), placental growth factor (PlGF), and sFlt-1/PlGF ratio in maternal serum and protein/creatinine ratio in urine sample to define the best cutoff point of these tests to discriminate between the patients with gestational hypertension and the patients with pre-eclampsia, to evaluate the possibility of using them as diagnostic methods. METHODS A prospective longitudinal study was performed, and blood samples were collected from 95 pregnant patients with hypertension to measure serum concentrations of biomarkers sFlt-1 and PlGF. Urine samples were collected for protein screening. Significance was set as p < 0.05. RESULTS The sFlt-1/PlGF ratio demonstrated a sensitivity of 57.5% and a specificity of 60% using 50.4 as a cutoff point. The test that showed the best accuracy in the diagnosis of pre-eclampsia was protein/creatinine ratio, with a sensitivity of 78.9% and a specificity of 70% using 0.4 as a cutoff point and showing an area under the receiver operating characteristic curve of 0.80 (p < 0.001). CONCLUSION No studied laboratory test proved to be fairly accurate for the diagnosis of pre-eclampsia, except for the protein/creatinine ratio. The evidence is insufficient to recommend biomarkers sFlt-1 and PlGF to be used for the diagnosis of pre-eclampsia.
Collapse
Affiliation(s)
- Catherine Primo Nogueira de Sá
- Department of Gynecology and Obstetrics, Universidade Federal de Ciências da Saúde, Porto Alegre, RS, Brazil.,Obstetrics Service, Santa Casa de Misericórdia, Porto Alegre, RS, Brazil.,Obstetrics Service, Hospital Moinhos de Vento, Porto Alegre, RS, Brazil
| | - Mirela Foresti Jiménez
- Department of Gynecology and Obstetrics, Universidade Federal de Ciências da Saúde, Porto Alegre, RS, Brazil.,Obstetrics Service, Santa Casa de Misericórdia, Porto Alegre, RS, Brazil
| | | | - Ellen Machado Arlindo
- Department of Gynecology and Obstetrics, Universidade Federal de Ciências da Saúde, Porto Alegre, RS, Brazil.,Obstetrics Service, Santa Casa de Misericórdia, Porto Alegre, RS, Brazil.,Obstetrics Service, Hospital Moinhos de Vento, Porto Alegre, RS, Brazil
| | - Antonio Celso Koehler Ayub
- Department of Gynecology and Obstetrics, Universidade Federal de Ciências da Saúde, Porto Alegre, RS, Brazil.,Obstetrics Service, Santa Casa de Misericórdia, Porto Alegre, RS, Brazil
| | - Rodrigo Bernardes Cardoso
- Department of Gynecology and Obstetrics, Universidade Federal de Ciências da Saúde, Porto Alegre, RS, Brazil.,Obstetrics Service, Santa Casa de Misericórdia, Porto Alegre, RS, Brazil
| | - Régis Kreitchmann
- Department of Gynecology and Obstetrics, Universidade Federal de Ciências da Saúde, Porto Alegre, RS, Brazil.,Obstetrics Service, Santa Casa de Misericórdia, Porto Alegre, RS, Brazil
| | - Patrícia El Beitune
- Department of Gynecology and Obstetrics, Universidade Federal de Ciências da Saúde, Porto Alegre, RS, Brazil.,Obstetrics Service, Santa Casa de Misericórdia, Porto Alegre, RS, Brazil
| |
Collapse
|
31
|
Szabo S, Karaszi K, Romero R, Toth E, Szilagyi A, Gelencser Z, Xu Y, Balogh A, Szalai G, Hupuczi P, Hargitai B, Krenacs T, Hunyadi-Gulyas E, Darula Z, Kekesi KA, Tarca AL, Erez O, Juhasz G, Kovalszky I, Papp Z, Than NG. Proteomic identification of Placental Protein 1 (PP1), PP8, and PP22 and characterization of their placental expression in healthy pregnancies and in preeclampsia. Placenta 2020; 99:197-207. [PMID: 32747003 PMCID: PMC8314955 DOI: 10.1016/j.placenta.2020.05.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Placental Protein 1 (PP1), PP8, and PP22 were isolated from the placenta. Herein, we aimed to identify PP1, PP8, and PP22 proteins and their placental and trophoblastic expression patterns to reveal potential involvement in pregnancy complications. METHODS We analyzed PP1, PP8, and PP22 proteins with LC-MS. We compared the placental behaviors of PP1, PP8, and PP22 to the predominantly placenta-expressed PP5/TFPI-2. Placenta-specificity scores were generated from microarray data. Trophoblasts were isolated from healthy placentas and differentiated; total RNA was isolated and subjected to microarray analysis. We assigned the placentas to the following groups: preterm controls, early-onset preeclampsia, early-onset preeclampsia with HELLP syndrome, term controls, and late-onset preeclampsia. After histopathologic examination, placentas were used for tissue microarray construction, immunostaining with anti-PP1, anti-PP5, anti-PP8, or anti-PP22 antibodies, and immunoscoring. RESULTS PP1, PP8, and PP22 were identified as 'nicotinate-nucleotide pyrophosphorylase', 'serpin B6', and 'protein disulfide-isomerase', respectively. Genes encoding PP1, PP8, and PP22 are not predominantly placenta-expressed, in contrast with PP5. PP1, PP8, and PP22 mRNA expression levels did not increase during trophoblast differentiation, in contrast with PP5. PP1, PP8, and PP22 immunostaining were detected primarily in trophoblasts, while PP5 expression was restricted to the syncytiotrophoblast. The PP1 immunoscore was higher in late-onset preeclampsia, while the PP5 immunoscore was higher in early-onset preeclampsia. DISCUSSION PP1, PP8, and PP22 are expressed primarily in trophoblasts but do not have trophoblast-specific regulation or functions. The distinct dysregulation of PP1 and PP5 expression in either late-onset or early-onset preeclampsia reflects different pathophysiological pathways in these preeclampsia subsets.
Collapse
Affiliation(s)
- Szilvia Szabo
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary; Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary.
| | - Katalin Karaszi
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary; First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary.
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, MI, USA; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA; Detroit Medical Center, Detroit, MI, USA; Department of Obstetrics and Gynecology, Florida International University, Miami, FL, USA
| | - Eszter Toth
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Andras Szilagyi
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Zsolt Gelencser
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Yi Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Andrea Balogh
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Gabor Szalai
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Petronella Hupuczi
- Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary
| | - Beata Hargitai
- West Midlands Perinatal Pathology Centre, Cellular Pathology Department, Birmingham Women's and Children's NHS FT, Birmingham, United Kingdom
| | - Tibor Krenacs
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | | | - Zsuzsanna Darula
- Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Katalin A Kekesi
- Department of Physiology and Neurobiology, ELTE Eotvos Lorand University, Budapest, Hungary; Laboratory of Proteomics, Institute of Biology, ELTE Eotvos Lorand University, Budapest, Hungary
| | - Adi L Tarca
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA
| | - Offer Erez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Maternity Department "D," Division of Obstetrics and Gynecology, Soroka University Medical Center, School of Medicine, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Gabor Juhasz
- Laboratory of Proteomics, Institute of Biology, ELTE Eotvos Lorand University, Budapest, Hungary; CRU Hungary Ltd., God, Hungary
| | - Ilona Kovalszky
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Zoltan Papp
- Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary
| | - Nandor Gabor Than
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary; First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary.
| |
Collapse
|
32
|
Langston-Cox A, Leo CH, Tare M, Wallace EM, Marshall SA. Sulforaphane improves vascular reactivity in mouse and human arteries after "preeclamptic-like" injury. Placenta 2020; 101:242-250. [PMID: 33032098 DOI: 10.1016/j.placenta.2020.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/16/2020] [Accepted: 09/01/2020] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The widespread maternal endothelial dysfunction that underlies the manifestations of preeclampsia is thought to arise from excessive placental production of antiangiogenic factors and enhanced oxidative stress. Therefore, we assessed whether the natural antioxidant sulforaphane could improve vascular function. METHODS Cell viability of human umbilical vein endothelial cells (HUVECs) was assessed after 24 or 48 h in normoxia (20% O2) or hypoxia (1% O2) with or without sulforaphane. To model vascular dysfunction associated with preeclampsia, mouse mesenteric arteries were incubated in trophoblast conditioned media (TCM), and human omental arteries incubated in preeclamptic explant media (PEM) with or without sulforaphane. Both media are rich in antiangiogenic compounds associated with preeclampsia. TCM was generated from primary cytotrophoblast cells from term placentae of normotensive, while PEM was generated from explants from preeclamptic women. Reactivity was assessed by wire myography. sulforaphane's actions as a vasodilator were also investigated. RESULTS Under conditions of hypoxia, sulforaphane improved HUVEC viability. In mouse mesenteric arteries, sulforaphane reduced contraction evoked by potassium (p < 0.001), phenylephrine and endothelin 1 (all p < 0.001). Sulforaphane also inhibited Ca2+-induced contraction (p = 0.014). Sulforaphane prevented TCM-induced augmentation of phenylephrine and angiotensin II-mediated contraction of mouse mesenteric arteries. In human omental arteries, sulforaphane induced vasodilation (p < 0.001), and prevented PEM-induced endothelial dysfunction by restoring arterial sensitivity to the endothelium-dependent vasodilator bradykinin (p = 0.008). DISCUSSION Sulforaphane causes relaxation in arteries and protects against arterial dysfunction induced by placental-derived antiangiogenic factors, which are known to contribute to the preeclampsia.
Collapse
Affiliation(s)
- A Langston-Cox
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - C H Leo
- Science, Mathematics and Technology, Singapore University of Technology & Design, Singapore
| | - M Tare
- Monash Rural Health, Monash University, Churchill, VIC, 3842, Australia; Department of Physiology, Monash University, Clayton, Australia
| | - E M Wallace
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - S A Marshall
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
33
|
Langston-Cox A, Muccini AM, Marshall SA, Yap, Palmer KR, Wallace EM, Ellery SJ. Sulforaphane improves syncytiotrophoblast mitochondrial function after in vitro hypoxic and superoxide injury. Placenta 2020; 96:44-54. [PMID: 32560857 DOI: 10.1016/j.placenta.2020.05.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/22/2020] [Accepted: 05/10/2020] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Placental mitochondrial dysfunction contributes to the oxidative stress that underlies preeclampsia. Here, we assessed whether sulforaphane (SFN) could improve syncytiotrophoblast mitochondrial function after in vitro hypoxic and superoxide injury. METHODS Placental cytotrophoblasts were isolated from healthy term placentae (n = 12) and incubated for 48 h in 8% O2 ± 1 μM SFN before acute (4hrs) or chronic (24hrs) hypoxic (1% O2), or superoxide (xanthine/xanthine oxidase) injury. Cytotrophoblasts were also isolated from preeclamptic placentae (n = 5) and cultured in 8% O2 ± 1 μM SFN. Mitochondrial respiration was measured using the Seahorse MitoStress XF assay. Cells were stained with mitotracker red to assess mitochondrial membrane health and mitochondrial gene expression assessed using RT-qPCR. RESULTS SFN prevented significant reductions in syncytiotrophoblast mitochondrial maximal respiration, spare respiratory capacity, basal respiration and ATP production following acute hypoxia. Chronic hypoxia only reduced maximal and spare respiratory capacity. SFN prevented these negative changes and increased respiration overall. Alternatively, acute superoxide injury significantly increased mitochondrial maximal respiration and spare respiratory capacity. SFN treatment further increased basal respiration following superoxide injury and prevented significant decreases in ATP production and coupling efficiency. In preeclamptic placentae, SFN significantly increased mitochondrial maximal respiration, spare respiratory capacity, basal respiration and ATP production, and decreased proton leak. SFN up-regulated mRNA expression of mitochondrial complexes and corrected an up-regulation in fission gene expression observed after hypoxic-superoxide injury. Finally, preliminary results suggest SFN prevented hypoxia-induced impairment of mitochondrial membrane structure. DISCUSSION SFN mitigated hypoxia and superoxide induced changes to syncytiotrophoblast mitochondrial function in vitro, and improved mitochondrial respiration in trophoblast cells from preeclamptic placentae.
Collapse
Affiliation(s)
- A Langston-Cox
- The Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia.
| | - A M Muccini
- The Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia; Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - S A Marshall
- The Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Yap
- The Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia; Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - K R Palmer
- The Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia; Monash Women's, Monash Health, Clayton, VIC, Australia
| | - E M Wallace
- The Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - S J Ellery
- The Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia; Hudson Institute of Medical Research, Clayton, VIC, Australia
| |
Collapse
|
34
|
Dickson MA, Peterson N, McRae KE, Pudwell J, Tayade C, Smith GN. Carbon monoxide increases utero-placental angiogenesis without impacting pregnancy specific adaptations in mice. Reprod Biol Endocrinol 2020; 18:49. [PMID: 32408878 PMCID: PMC7227344 DOI: 10.1186/s12958-020-00594-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 04/22/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cigarette smokers have a reduced risk of developing preeclampsia, possibly attributed to an increase in carbon monoxide (CO) levels. Carbon monoxide is a gasotransmitter that has been implicated in maintaining vascular tone, increasing angiogenesis, and reducing inflammation and apoptosis at physiological concentrations. Moderately increasing CO concentrations may have therapeutic potential to prevent or treat preeclampsia; however, the effects of CO on pregnancy are under studied. Our objective was to investigate the effect of CO on major angiogenic and inflammatory markers in pregnancy, and to evaluate the effect of CO on indicators of placental health. FINDINGS Pregnant CD-1 mice were constantly exposed to either ambient air or 250 ppm CO from conception until gestation day (GD)10.5 or GD16.5. Using a qRT-PCR array, we identified that CO increased expression of major angiogenic genes at the implantation site on GD10.5, but not GD16.5. Pro-inflammatory cytokines in the plasma and tissue lysates from implantation sites in treated mice were not significantly different compared to controls. Additionally, CO did not alter the implantation site phenotype, in terms of proliferative capacity, invasiveness of trophoblasts, or abundance of uterine natural killer cells. CONCLUSIONS This study suggests that CO exposure is pro-angiogenic at the maternal-fetal interface, and is not associated with demonstrable concerns during murine pregnancy. Future studies are required to validate safety and efficacy of CO as a potential therapeutic for vascular insufficiency diseases such as preeclampsia and intrauterine growth restriction.
Collapse
Affiliation(s)
- Megan A. Dickson
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6 Canada
| | - Nichole Peterson
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6 Canada
| | - Karalyn E. McRae
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6 Canada
| | - Jessica Pudwell
- Department of Obstetrics and Gynaecology, Queen’s University, Kingston Health Sciences Centre, 76 Stuart St, Kingston, K7L 2V7 Canada
| | - Chandrakant Tayade
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6 Canada
| | - Graeme N. Smith
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6 Canada
- Department of Obstetrics and Gynaecology, Queen’s University, Kingston Health Sciences Centre, 76 Stuart St, Kingston, K7L 2V7 Canada
| |
Collapse
|
35
|
Hitzerd E, Broekhuizen M, Neuman RI, Colafella KMM, Merkus D, Schoenmakers S, Simons SHP, Reiss IKM, Danser AHJ. Human Placental Vascular Reactivity in Health and Disease: Implications for the Treatment of Pre-eclampsia. Curr Pharm Des 2020; 25:505-527. [PMID: 30950346 DOI: 10.2174/1381612825666190405145228] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/29/2019] [Indexed: 12/17/2022]
Abstract
Adequate development of the placenta is essential for optimal pregnancy outcome. Pre-eclampsia (PE) is increasingly recognized to be a consequence of placental dysfunction and can cause serious maternal and fetal complications during pregnancy. Furthermore, PE increases the risk of neonatal problems and has been shown to be a risk factor for cardiovascular disease of the mother later in life. Currently, there is no adequate treatment for PE, mainly because its multifactorial pathophysiology remains incompletely understood. It originates in early pregnancy with abnormal placentation and involves a cascade of dysregulated systems in the placental vasculature. To investigate therapeutic strategies it is essential to understand the regulation of vascular reactivity and remodeling of blood vessels in the placenta. Techniques using human tissue such as the ex vivo placental perfusion model provide insight in the vasoactive profile of the placenta, and are essential to study the effects of drugs on the fetal vasculature. This approach highlights the different pathways that are involved in the vascular regulation of the human placenta, changes that occur during PE and the importance of focusing on restoring these dysfunctional systems when studying treatment strategies for PE.
Collapse
Affiliation(s)
- Emilie Hitzerd
- Department of Pediatrics, Division of Neonatology, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Department of Internal Medicine; Division of Pharmacology and Vascular Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Michelle Broekhuizen
- Department of Pediatrics, Division of Neonatology, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Department of Internal Medicine; Division of Pharmacology and Vascular Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Department of Cardiology; Division of Experimental Cardiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Rugina I Neuman
- Department of Internal Medicine; Division of Pharmacology and Vascular Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Department of Gynecology and Obstetrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Katrina M Mirabito Colafella
- Department of Internal Medicine; Division of Pharmacology and Vascular Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Cardiovascular Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.,Department of Physiology, Monash University, Melbourne, Australia
| | - Daphne Merkus
- Department of Cardiology; Division of Experimental Cardiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Sam Schoenmakers
- Department of Gynecology and Obstetrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Sinno H P Simons
- Department of Pediatrics, Division of Neonatology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Irwin K M Reiss
- Department of Pediatrics, Division of Neonatology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - A H Jan Danser
- Department of Internal Medicine; Division of Pharmacology and Vascular Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
36
|
Oxidative stress: Normal pregnancy versus preeclampsia. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165354. [DOI: 10.1016/j.bbadis.2018.12.005] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/22/2018] [Accepted: 12/05/2018] [Indexed: 02/03/2023]
|
37
|
Gallardo V, González M, Toledo F, Sobrevia L. Role of heme oxygenase 1 and human chorionic gonadotropin in pregnancy associated diseases. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165522. [DOI: 10.1016/j.bbadis.2019.07.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 07/25/2019] [Accepted: 07/27/2019] [Indexed: 01/13/2023]
|
38
|
Iriyama T, Wang W, Parchim NF, Sayama S, Kumasawa K, Nagamatsu T, Song A, Xia Y, Kellems RE. Reciprocal upregulation of hypoxia-inducible factor-1α and persistently enhanced placental adenosine signaling contribute to the pathogenesis of preeclampsia. FASEB J 2020; 34:4041-4054. [PMID: 31930569 DOI: 10.1096/fj.201902583r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/28/2019] [Accepted: 12/30/2019] [Indexed: 02/02/2023]
Abstract
Recent evidence indicates that elevated placental adenosine signaling contributes to preeclampsia (PE). However, the molecular basis for the chronically enhanced placental adenosine signaling in PE remains unclear. Here, we report that hypoxia-inducible factor-1α (HIF-1α) is crucial for the enhancement of placental adenosine signaling. Utilizing a pharmacologic approach to reduce placental adenosine levels, we found that enhanced adenosine underlies increased placental HIF-1α in an angiotensin receptor type 1 receptor agonistic autoantibody (AT1 -AA)-induced mouse model of PE. Knockdown of placental HIF-1α in vivo suppressed the accumulation of adenosine and increased ecto-5'-nucleotidase (CD73) and adenosine A2B receptor (ADORA2B) in the placentas of PE mouse models induced by AT1 -AA or LIGHT, a TNF superfamily cytokine (TNFSF14). Human in vitro studies using placental villous explants demonstrated that increased HIF-1α resulting from ADORA2B activation facilitates the induction of CD73, ADORA2B, and FLT-1 expression. Overall, we demonstrated that (a) elevated placental HIF-1α by AT1 -AA or LIGHT upregulates CD73 and ADORA2B expression and (b) enhanced adenosine signaling through upregulated ADORA2B induces placental HIF-1α expression, which creates a positive feedback loop that promotes FLT-1 expression leading to disease development. Our results suggest that adenosine-based therapy targeting the malicious cycle of placental adenosine signaling may elicit therapeutic effects on PE.
Collapse
Affiliation(s)
- Takayuki Iriyama
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Wei Wang
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, TX, USA.,Department of Nephrology, Xiangya Hospital of Central South University, Changsha, P.R. China
| | - Nicholas F Parchim
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, TX, USA.,Department of Emergency Medicine, The University of New Mexico Hospital, Albuquerque, NM, USA
| | - Seisuke Sayama
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Keiichi Kumasawa
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Anren Song
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, TX, USA.,Department of Nephrology, Xiangya Hospital of Central South University, Changsha, P.R. China.,Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Rodney E Kellems
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, TX, USA.,Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
39
|
Kovtun O, Tsyvian P. Developmental origins of accelerated cardiovascular aging. BIO WEB OF CONFERENCES 2020. [DOI: 10.1051/bioconf/20202201002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The Barker hypothesis of fetal origins of disease, derived from observational epidemiological studies, has been modified by the realization that the early influences on cardiovascular development may be recognized in the fetus. Aging and developmental programming are both associated with oxidative stress and endothelial dysfunction, suggesting common mechanistic origins. Preeclampsia (PE) is the utmost variant of placental insufficiency and is associated with an increase of serum concentrations of inflammation and antiangiogenic agents in mother and fetus. All these factors are specific for the endothelial dysfunction and able to trigger the epigenetic programming of cardiovascular diseases and accelerated aging. The concentrations of cytokines, tumor necrosis factor, C-reactive protein, NO metabolites, endothelin-1 and homocysteine were measured in serum of women with a normal pregnancy (n=27), PE (n=30) and their newborn infants. The concentrations of all these factors were higher in serum of women and newborns of PE group than in serum of women and newborns of the normal group. The significantly positive correlation between factors concentrations in maternal and newborn’s serum was demonstrated in all groups. We suggest the common source of these agents to be the syncytiotrophoblast cells contacting with both maternal and fetal blood and play a significant role in intrauterine programming and epigenetic triggering of accelerated cardiovascular aging.
Collapse
|
40
|
Deepak V, Sahu MB, Yu J, Jones JW, Kane MA, Taylor RN, Badell ML, Sidell N, Rajakumar A. Retinoic Acid Is a Negative Regulator of sFLT1 Expression in Decidual Stromal Cells, and Its Levels Are Reduced in Preeclamptic Decidua. Hypertension 2019; 73:1104-1111. [PMID: 30879360 DOI: 10.1161/hypertensionaha.118.12564] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
sFLT1 (soluble VEGF [vascular endothelial growth factor] receptor-1) levels are increased in preeclampsia-a pathological condition of pregnancy. The mechanism of sFLT1 overexpression by gestational tissues, particularly the decidua, remains unknown. Mass spectrometry measurement of the active retinoid metabolite, all-trans retinoic acid (RA), showed significantly lower levels of RA in preeclamptic versus normotensive decidua. In this study, we investigated the involvement of RA in regulating decidual sFLT1 expression. When decidual stromal cells (DSCs) isolated from the decidua basalis of normotensive and preeclampsia placentas were treated with BMS493-a pan-RAR (RA nuclear receptor) antagonist-upregulation of sFLT1 expression was observed. Conversely, treatment with RA resulted in downregulation of sFLT1 in normotensive DSCs and preeclampsia DSCs. Unlike treatment with cAMP, which induces decidualization while downregulating sFLT1, RA treatment did not alter DSC expression of prolactin-a marker of decidualization-or FOXO1 (forkhead box protein 01)-a transcription factor required for prolactin upregulation. TFAP2A (transcription factor AP-2-alpha [activating enhancer-binding protein 2 alpha]), a different transcription factor was upregulated in normotensive DSCs but not in preeclampsia DSCs after RA treatment. Collectively, our data show that RA suppresses sFLT1 expression in DSCs independently of cellular decidualization. These findings suggest that reduced decidual RA levels may contribute to preeclampsia pathogenesis by allowing sFLT1 accumulation at the maternal-fetal interface.
Collapse
Affiliation(s)
- Venkataraman Deepak
- From the Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA (V.D., M.B.S., M.L.B., N.S., A.R.)
| | - Margaret B Sahu
- From the Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA (V.D., M.B.S., M.L.B., N.S., A.R.)
| | - Jianshi Yu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore (J.Y., J.W.J., M.A.K.)
| | - Jace W Jones
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore (J.Y., J.W.J., M.A.K.)
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore (J.Y., J.W.J., M.A.K.)
| | - Robert N Taylor
- Department of Obstetrics and Gynecology, University of Utah School of Medicine, Salt Lake City (R.N.T.)
| | - Martina L Badell
- From the Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA (V.D., M.B.S., M.L.B., N.S., A.R.)
| | - Neil Sidell
- From the Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA (V.D., M.B.S., M.L.B., N.S., A.R.)
| | - Augustine Rajakumar
- From the Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA (V.D., M.B.S., M.L.B., N.S., A.R.)
| |
Collapse
|
41
|
Padayachee S, Moodley J, Naicker T. A Review of Angiogenic Imbalance in HIV-Infected Hypertensive Disorders of Pregnancy. Curr Hypertens Rep 2019; 21:69. [PMID: 31342170 DOI: 10.1007/s11906-019-0970-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW This review provides a comprehensive insight into the angiogenic profile of hypertensive and normotensive pregnancies compromised by HIV infection. Furthermore, we evaluate the economic implementation of the sFlt-1/PlGF ratio and review the reports on therapeutic apheresis in limiting sFlt-1 production. RECENT FINDINGS In preeclampsia, an increased expression of sFlt-1 triggers angiogenic imbalance. Women of African ancestry have high levels of angiogenic factors than other racial groups. The sFlt-1/PlGF ratio shows promise in the early assessment of preeclampsia, while sFlt-1 apheresis restores angiogenic imbalance. Studies suggest antiretroviral therapy does not impact the angiogenic shift in preeclampsia development. The angiogenic profile in pregnant women of different races influences preeclampsia development. Despite the opposing immune response in HIV infection and preeclampsia, the HIV tat protein strongly mimics vascular endothelial growth factor (VEGF); hence, it is plausible to assume that HIV infection may ameliorate the angiogenic imbalance in preeclampsia.
Collapse
MESH Headings
- Angiogenic Proteins/blood
- Angiogenic Proteins/physiology
- Biomarkers/blood
- Biomarkers/metabolism
- Blood Component Removal
- Female
- HIV Infections/blood
- HIV Infections/complications
- HIV Infections/physiopathology
- Humans
- Hypertension, Pregnancy-Induced/blood
- Hypertension, Pregnancy-Induced/physiopathology
- Hypertension, Pregnancy-Induced/therapy
- Membrane Proteins/blood
- Membrane Proteins/physiology
- Neovascularization, Pathologic/blood
- Neovascularization, Pathologic/physiopathology
- Neovascularization, Physiologic/physiology
- Pre-Eclampsia/blood
- Pre-Eclampsia/physiopathology
- Pre-Eclampsia/therapy
- Pregnancy
- Pregnancy Complications, Infectious/blood
- Pregnancy Complications, Infectious/physiopathology
- Vascular Endothelial Growth Factor A/blood
- Vascular Endothelial Growth Factor A/physiology
- Vascular Endothelial Growth Factor Receptor-1/blood
- Vascular Endothelial Growth Factor Receptor-1/physiology
- tat Gene Products, Human Immunodeficiency Virus/blood
- tat Gene Products, Human Immunodeficiency Virus/physiology
Collapse
Affiliation(s)
- Sayuri Padayachee
- Optics and Imaging Centre, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Private Bag 7, Congella, Durban, KwaZulu-Natal, 4013, South Africa.
| | - Jagidesa Moodley
- Women's Health and HIV Research Group, Department of Obstetrics and Gynecology, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Private Bag 7, Congella, Durban, KwaZulu-Natal, 4013, South Africa
| |
Collapse
|
42
|
Tong S, Joy Kaitu'u-Lino T, Walker SP, MacDonald TM. Blood-based biomarkers in the maternal circulation associated with fetal growth restriction. Prenat Diagn 2019; 39:947-957. [PMID: 31299098 DOI: 10.1002/pd.5525] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/28/2019] [Accepted: 07/03/2019] [Indexed: 11/11/2022]
Abstract
Fetal growth restriction (FGR) is associated with threefold to fourfold increased risk of stillbirth. Identifying FGR, through its commonly used surrogate-the small-for-gestational-age (SGA, estimated fetal weight and/or abdominal circumference <10th centile) fetus-and instituting fetal surveillance and timely delivery decrease stillbirth risk. Methods available to clinicians for antenatal identification of SGA fetuses have surprisingly poor sensitivity. About 80% of cases remain undetected. Measuring the symphysis-fundal height detects only 20% of SGA fetuses, and even universal third trimester ultrasound detects, at best, 57% of those born SGA. There is an urgent need to find better ways to identify this at-risk cohort. This review summarises efforts to identify molecular biomarkers (proteins, metabolites, or ribonucleic acids) that could be used to better predict FGR. Most studies examining potential biomarkers to date have utilised case-control study designs without proceeding to validation in independent cohorts. To develop a robust test for FGR, large prospective studies are required with a priori validation plans and cohorts. Given that current clinical care detects 20% of SGA fetuses, even a screening test with ≥60% sensitivity at 90% specificity could be clinically useful, if developed. This may be an achievable aspiration. If discovered, such a test may decrease stillbirth.
Collapse
Affiliation(s)
- Stephen Tong
- Mercy Perinatal, Mercy Hospital for Women, Melbourne, Victoria, Australia.,Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia
| | - Tu'uhevaha Joy Kaitu'u-Lino
- Mercy Perinatal, Mercy Hospital for Women, Melbourne, Victoria, Australia.,Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia
| | - Susan Philippa Walker
- Mercy Perinatal, Mercy Hospital for Women, Melbourne, Victoria, Australia.,Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia
| | - Teresa Mary MacDonald
- Mercy Perinatal, Mercy Hospital for Women, Melbourne, Victoria, Australia.,Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
43
|
Trapiella-Alfonso L, Alexandre L, Fraichard C, Pons K, Dumas S, Huart L, Gaucher JF, Hebert-Schuster M, Guibourdenche J, Fournier T, Vidal M, Broutin I, Lecomte-Raclet L, Malaquin L, Descroix S, Tsatsaris V, Gagey-Eilstein N, Lecarpentier E. VEGF (Vascular Endothelial Growth Factor) Functionalized Magnetic Beads in a Microfluidic Device to Improve the Angiogenic Balance in Preeclampsia. Hypertension 2019; 74:145-153. [DOI: 10.1161/hypertensionaha.118.12380] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Laura Trapiella-Alfonso
- From the UMR 8638 (L.T.-A., K.P., L.H., M.V., N.G.-E.), CNRS, Université Paris Descartes, Faculté des Sciences Pharmaceutiques et Biologiques, Sorbonne Paris Cité, Paris, France
| | - Lucile Alexandre
- UMR 168 CNRS Institut Curie, PSL Research University, Institut Pierre Gilles de Gennes, Paris, France (L.A., S.Dumas, S. Descroix.)
| | - Camille Fraichard
- Cochin Hospital, Assistance Publique-Hôpital de Paris, DHU Risques et grossesse, Paris Descartes University, INSERM UMR 1139, Fondation PremUp, Paris, France (C.F., M.H.-S., J.G., T.F., V.T.)
| | - Kelly Pons
- From the UMR 8638 (L.T.-A., K.P., L.H., M.V., N.G.-E.), CNRS, Université Paris Descartes, Faculté des Sciences Pharmaceutiques et Biologiques, Sorbonne Paris Cité, Paris, France
- Centre Hospitalier Intercommunal de Créteil, Service de Gynécologie Obstétrique, France (K.P., E.L.)
| | - Simon Dumas
- UMR 168 CNRS Institut Curie, PSL Research University, Institut Pierre Gilles de Gennes, Paris, France (L.A., S.Dumas, S. Descroix.)
| | - Lucie Huart
- From the UMR 8638 (L.T.-A., K.P., L.H., M.V., N.G.-E.), CNRS, Université Paris Descartes, Faculté des Sciences Pharmaceutiques et Biologiques, Sorbonne Paris Cité, Paris, France
- UMR 8038 (L.H., J.-F.G., I.B.), CNRS, Université Paris Descartes, Faculté des Sciences Pharmaceutiques et Biologiques, Sorbonne Paris Cité, Paris, France
| | - Jean-François Gaucher
- UMR 8038 (L.H., J.-F.G., I.B.), CNRS, Université Paris Descartes, Faculté des Sciences Pharmaceutiques et Biologiques, Sorbonne Paris Cité, Paris, France
| | - Marylise Hebert-Schuster
- Cochin Hospital, Assistance Publique-Hôpital de Paris, DHU Risques et grossesse, Paris Descartes University, INSERM UMR 1139, Fondation PremUp, Paris, France (C.F., M.H.-S., J.G., T.F., V.T.)
- Cochin Hospital, UF d’Hormonologie, Service de Biologie Automatisée, Paris, France (M.H.-S., J.G.)
| | - Jean Guibourdenche
- Cochin Hospital, Assistance Publique-Hôpital de Paris, DHU Risques et grossesse, Paris Descartes University, INSERM UMR 1139, Fondation PremUp, Paris, France (C.F., M.H.-S., J.G., T.F., V.T.)
- Cochin Hospital, UF d’Hormonologie, Service de Biologie Automatisée, Paris, France (M.H.-S., J.G.)
| | - Thierry Fournier
- Cochin Hospital, Assistance Publique-Hôpital de Paris, DHU Risques et grossesse, Paris Descartes University, INSERM UMR 1139, Fondation PremUp, Paris, France (C.F., M.H.-S., J.G., T.F., V.T.)
| | - Michel Vidal
- From the UMR 8638 (L.T.-A., K.P., L.H., M.V., N.G.-E.), CNRS, Université Paris Descartes, Faculté des Sciences Pharmaceutiques et Biologiques, Sorbonne Paris Cité, Paris, France
| | - Isabelle Broutin
- UMR 8038 (L.H., J.-F.G., I.B.), CNRS, Université Paris Descartes, Faculté des Sciences Pharmaceutiques et Biologiques, Sorbonne Paris Cité, Paris, France
| | - Laurence Lecomte-Raclet
- URC - CIC P1419, Cochin Hôtel-Dieu Hospital, Assistance Publique–Hôpitaux de Paris, France (L.L.-R.)
| | | | - Stéphanie Descroix
- UMR 168 CNRS Institut Curie, PSL Research University, Institut Pierre Gilles de Gennes, Paris, France (L.A., S.Dumas, S. Descroix.)
| | - Vassilis Tsatsaris
- Cochin Hospital, Assistance Publique-Hôpital de Paris, DHU Risques et grossesse, Paris Descartes University, INSERM UMR 1139, Fondation PremUp, Paris, France (C.F., M.H.-S., J.G., T.F., V.T.)
| | - Nathalie Gagey-Eilstein
- From the UMR 8638 (L.T.-A., K.P., L.H., M.V., N.G.-E.), CNRS, Université Paris Descartes, Faculté des Sciences Pharmaceutiques et Biologiques, Sorbonne Paris Cité, Paris, France
| | - Edouard Lecarpentier
- Université Paris Est Créteil, France (E.L.)
- INSERM, U955, Institut Mondor de Recherche Biomédicale, Créteil, France (E.L.)
- Centre Hospitalier Intercommunal de Créteil, Service de Gynécologie Obstétrique, France (K.P., E.L.)
| |
Collapse
|
44
|
Maternal preeclampsia and the risk of pediatric gastrointestinal diseases of the offspring: A population-based cohort study. Pregnancy Hypertens 2019; 17:144-147. [DOI: 10.1016/j.preghy.2019.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 05/30/2019] [Accepted: 06/18/2019] [Indexed: 11/21/2022]
|
45
|
Balogh A, Toth E, Romero R, Parej K, Csala D, Szenasi NL, Hajdu I, Juhasz K, Kovacs AF, Meiri H, Hupuczi P, Tarca AL, Hassan SS, Erez O, Zavodszky P, Matko J, Papp Z, Rossi SW, Hahn S, Pallinger E, Than NG. Placental Galectins Are Key Players in Regulating the Maternal Adaptive Immune Response. Front Immunol 2019; 10:1240. [PMID: 31275299 PMCID: PMC6593412 DOI: 10.3389/fimmu.2019.01240] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/16/2019] [Indexed: 12/12/2022] Open
Abstract
Galectins are potent immunomodulators that regulate maternal immune responses in pregnancy and prevent the rejection of the semi-allogeneic fetus that also occurs in miscarriages. We previously identified a gene cluster on Chromosome 19 that expresses a subfamily of galectins, including galectin-13 (Gal-13) and galectin-14 (Gal-14), which emerged in anthropoid primates. These galectins are expressed only by the placenta and induce the apoptosis of activated T lymphocytes, possibly contributing to a shifted maternal immune balance in pregnancy. The placental expression of Gal-13 and Gal-14 is decreased in preeclampsia, a life-threatening obstetrical syndrome partly attributed to maternal anti-fetal rejection. This study is aimed at revealing the effects of Gal-13 and Gal-14 on T cell functions and comparing the expression of these galectins in placentas from healthy pregnancies and miscarriages. First-trimester placentas were collected from miscarriages and elective termination of pregnancies, tissue microarrays were constructed, and then the expression of Gal-13 and Gal-14 was analyzed by immunohistochemistry and immunoscoring. Recombinant Gal-13 and Gal-14 were expressed and purified, and their effects were investigated on primary peripheral blood T cells. The binding of Gal-13 and Gal-14 to T cells and the effects of these galectins on apoptosis, activation marker (CD25, CD71, CD95, HLA-DR) expression and cytokine (IL-1β, IL-6, IL-8, IL-10, IFNγ) production of T cells were examined by flow cytometry. Gal-13 and Gal-14 are primarily expressed by the syncytiotrophoblast at the maternal-fetal interface in the first trimester, and their placental expression is decreased in miscarriages compared to first-trimester controls. Recombinant Gal-13 and Gal-14 bind to T cells in a population- and activation-dependent manner. Gal-13 and Gal-14 induce apoptosis of Th and Tc cell populations, regardless of their activation status. Out of the investigated activation markers, Gal-14 decreases the cell surface expression of CD71, Gal-13 increases the expression of CD25, and both galectins increase the expression of CD95 on T cells. Non-activated T cells produce larger amounts of IL-8 in the presence of Gal-13 or Gal-14. In conclusion, these results show that Gal-13 and Gal-14 already provide an immunoprivileged environment at the maternal-fetal interface during early pregnancy, and their reduced expression is related to miscarriages.
Collapse
Affiliation(s)
- Andrea Balogh
- Systems Biology of Reproduction Momentum Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary.,Department of Immunology, Eotvos Lorand University, Budapest, Hungary
| | - Eszter Toth
- Systems Biology of Reproduction Momentum Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD and Detroit, MI, United States.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, United States.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, United States.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
| | - Katalin Parej
- Systems Biology of Reproduction Momentum Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary.,Structural Biophysics Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Diana Csala
- Systems Biology of Reproduction Momentum Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Nikolett L Szenasi
- Systems Biology of Reproduction Momentum Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Istvan Hajdu
- Structural Biophysics Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Kata Juhasz
- Systems Biology of Reproduction Momentum Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Arpad F Kovacs
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | | | - Petronella Hupuczi
- Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary
| | - Adi L Tarca
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD and Detroit, MI, United States.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States.,Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, United States
| | - Sonia S Hassan
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD and Detroit, MI, United States.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States.,Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Offer Erez
- Division of Obstetrics and Gynecology, Maternity Department "D", Faculty of Health Sciences, Soroka University Medical Center, School of Medicine, Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Peter Zavodszky
- Structural Biophysics Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Janos Matko
- Department of Immunology, Eotvos Lorand University, Budapest, Hungary
| | - Zoltan Papp
- Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary.,Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| | - Simona W Rossi
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Sinuhe Hahn
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Eva Pallinger
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Nandor Gabor Than
- Systems Biology of Reproduction Momentum Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary.,Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary.,First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
46
|
Mani C, Kochhar P, Ravikumar G, Dwarkanath P, Sheela CN, George S, Thomas A, Crasta J, Thomas T, Kurpad AV, Mukhopadhyay A. Placental expression of ENG, VEGF, and FLT: Gender-specific associations with maternal vitamin B 12 status. Eur J Clin Nutr 2019; 74:176-182. [PMID: 31209272 DOI: 10.1038/s41430-019-0449-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 03/03/2019] [Accepted: 06/03/2019] [Indexed: 11/09/2022]
Abstract
OBJECTIVES Adequate vitamin B12 is a requisite during pregnancy and its deficiency is linked with increased risk for adverse outcomes, likely mediated by impaired placental angiogenesis. Thus, we aimed to test associations of maternal vitamin B12 status with the placental expression of angiogenesis-associated genes ENG, VEGF, and FLT. SUBJECTS/METHODS In this retrospective case-control study, placental and maternal trimester 1 blood samples (n = 104) were collected from small for gestational age (SGA) and appropriate for gestational age (AGA) full-term singleton pregnancies. Maternal trimester 1 vitamin B12 status was measured. Placentae and neonates were weighed at birth. Realtime quantitative PCR was performed to assess placental transcript abundance of ENG, VEGF, and FLT normalized to a panel of reference genes. Associations of placental transcript abundance of the genes with maternal trimester 1 vitamin B12 status were evaluated. RESULTS Placental ENG transcript abundance associated negatively with maternal trimester 1 vitamin B12 status (β = -0.461, P = 0.017, n = 104). This association was specific to the female births (β = -0.590, P = 0.014, n = 60). Placental VEGF transcript levels were negatively associated with maternal trimester 1 vitamin B12 status only in the female births (β = -1.995, P = 0.029). Placental FLT transcript levels were not associated with maternal trimester 1 vitamin B12 status. CONCLUSION Maternal trimester 1 vitamin B12 status was associated negatively with placental ENG and VEGF expression predominantly in the female births. Therefore, we hypothesize that the placenta adapts to low maternal vitamin B12 status by up-regulating angiogenic pathways in a gender-specific manner.
Collapse
Affiliation(s)
- C Mani
- Division of Nutrition, St. John's Research Institute, St. John's Academy of Health Sciences, Bangalore, India
| | - P Kochhar
- Division of Nutrition, St. John's Research Institute, St. John's Academy of Health Sciences, Bangalore, India
| | - G Ravikumar
- Department of Pathology, St John's Medical College Hospital, Bangalore, India
| | - P Dwarkanath
- Division of Nutrition, St. John's Research Institute, St. John's Academy of Health Sciences, Bangalore, India
| | - C N Sheela
- Department of Obstetrics and Gynaecology, St John's Medical College Hospital, Bangalore, India
| | - S George
- Department of Obstetrics and Gynaecology, St John's Medical College Hospital, Bangalore, India
| | - A Thomas
- Department of Obstetrics and Gynaecology, St John's Medical College Hospital, Bangalore, India
| | - J Crasta
- Department of Pathology, St John's Medical College Hospital, Bangalore, India
| | - T Thomas
- Department of Biostatistics, St. John's Medical College, St. John's Academy of Health Sciences, Bangalore, India
| | - A V Kurpad
- Division of Nutrition, St. John's Research Institute, St. John's Academy of Health Sciences, Bangalore, India
| | - A Mukhopadhyay
- Division of Nutrition, St. John's Research Institute, St. John's Academy of Health Sciences, Bangalore, India.
| |
Collapse
|
47
|
Placental effects and transfer of sildenafil in healthy and preeclamptic conditions. EBioMedicine 2019; 45:447-455. [PMID: 31204276 PMCID: PMC6642075 DOI: 10.1016/j.ebiom.2019.06.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/28/2019] [Accepted: 06/06/2019] [Indexed: 12/17/2022] Open
Abstract
Background The phosphodiesterase-5 inhibitor (PDE5) sildenafil has emerged as a promising treatment for preeclampsia (PE). However, a sildenafil trial was recently halted due to lack of effect and increased neonatal morbidity. Methods Ex vivo dual-sided perfusion of an isolated cotyledon and wire-myography on chorionic plate arteries were performed to study the effects of sildenafil and the non-selective PDE inhibitor vinpocetine on the response to the NO donor sodium nitroprusside (SNP) under healthy and PE conditions. Ex vivo perfusion was also used to study placental transfer of sildenafil in 6 healthy and 2 PE placentas. Furthermore, placental mRNA and protein levels of eNOS, iNOS, PDE5 and PDE1 were quantified. Findings Sildenafil and vinpocetine significantly enhanced SNP responses in chorionic plate arteries of healthy, but not PE placentas. Only sildenafil acutely decreased baseline tension in arteries of both healthy and PE placentas. At steady state, the foetal-to-maternal transfer ratio of sildenafil was 0·37 ± 0·03 in healthy placentas versus 0·66 and 0·47 in the 2 PE placentas. mRNA and protein levels of PDE5, eNOS and iNOS were comparable in both groups, while PDE1 levels were lower in PE. Interpretation The absence of sildenafil-induced NO potentiation in arteries of PE placentas, combined with the non-PDE-mediated effects of sildenafil and the lack of PDE5 upregulation in PE, argue against sildenafil as the preferred drug of use in PE. Moreover, increased placental transfer of sildenafil in PE might underlie the neonatal morbidity in the STRIDER trial. Fund This study was funded by an mRACE Erasmus MC grant.
Collapse
|
48
|
Phipps EA, Thadhani R, Benzing T, Karumanchi SA. Pre-eclampsia: pathogenesis, novel diagnostics and therapies. Nat Rev Nephrol 2019; 15:275-289. [PMID: 30792480 PMCID: PMC6472952 DOI: 10.1038/s41581-019-0119-6] [Citation(s) in RCA: 601] [Impact Index Per Article: 100.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pre-eclampsia is a complication of pregnancy that is associated with substantial maternal and fetal morbidity and mortality. The disease presents with new-onset hypertension and often proteinuria in the mother, which can progress to multi-organ dysfunction, including hepatic, renal and cerebral disease, if the fetus and placenta are not delivered. Maternal endothelial dysfunction due to circulating factors of fetal origin from the placenta is a hallmark of pre-eclampsia. Risk factors for the disease include maternal comorbidities, such as chronic kidney disease, hypertension and obesity; a family history of pre-eclampsia, nulliparity or multiple pregnancies; and previous pre-eclampsia or intrauterine fetal growth restriction. In the past decade, the discovery and characterization of novel antiangiogenic pathways have been particularly impactful both in increasing understanding of the disease pathophysiology and in directing predictive and therapeutic efforts. In this Review, we discuss the pathogenic role of antiangiogenic proteins released by the placenta in the development of pre-eclampsia and review novel therapeutic strategies directed at restoring the angiogenic imbalance observed during pre-eclampsia. We also highlight other notable advances in the field, including the identification of long-term maternal and fetal risks conferred by pre-eclampsia.
Collapse
Affiliation(s)
- Elizabeth A Phipps
- Nephrology Division, Brigham and Women's Hospital, Boston, MA, USA
- Nephrology Division, Massachusetts General Hospital, Boston, MA, USA
| | - Ravi Thadhani
- Nephrology Division, Massachusetts General Hospital, Boston, MA, USA
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - S Ananth Karumanchi
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Nephrology Division, Departments of Medicine, Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
49
|
Karaszi K, Szabo S, Juhasz K, Kiraly P, Kocsis-Deak B, Hargitai B, Krenacs T, Hupuczi P, Erez O, Papp Z, Kovalszky I, Than NG. Increased placental expression of Placental Protein 5 (PP5) / Tissue Factor Pathway Inhibitor-2 (TFPI-2) in women with preeclampsia and HELLP syndrome: Relevance to impaired trophoblast invasion? Placenta 2019; 76:30-39. [PMID: 30803712 DOI: 10.1016/j.placenta.2019.01.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 01/09/2019] [Accepted: 01/11/2019] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Placental Protein 5 (PP5)/Tissue Factor Pathway Inhibitor-2 (TFPI-2) is an extracellular matrix-associated protein mainly expressed by the syncytiotrophoblast that may regulate trophoblast invasion. Our aim was to study placental PP5/TFPI-2 expression and its relation to placental pathology in various forms of preeclampsia and HELLP syndrome. METHODS Placental and maternal blood specimens were collected at the time of delivery from the same women in the following groups: 1) early controls; 2) early preeclampsia; 3) early preeclampsia with HELLP syndrome; 4) late controls; and 5) late preeclampsia. After histopathological examination, placental specimens were immunostained with polyclonal anti-PP5/TFPI-2 antibody on Western blot and tissue microarray immunohistochemistry. Placental PP5/TFPI-2 immunoscores were assessed manually and with a semi-automated method. Maternal sera were immunoassayed for PP5/TFPI-2. RESULTS PP5/TFPI-2 was localized to the cytoplasm of syncytiotrophoblast. Manual and semi-automated PP5/TFPI-2 immunoscores were higher in early preeclampsia with or without HELLP syndrome but not in late preeclampsia than in respective controls. In patients with preeclampsia, the correlation of placental PP5/TFPI-2 expression with maternal vascular malperfusion score of the placenta was positive while it was negative with birthweight and placental weight. Maternal serum PP5/TFPI-2 concentration was higher in early preeclampsia and it tended to be higher in early preeclampsia with HELLP syndrome than in early controls. DISCUSSION Our findings suggest that an increased placental PP5/TFPI-2 expression may be associated with abnormal placentation in early preeclampsia, with or without HELLP syndrome.
Collapse
Affiliation(s)
- Katalin Karaszi
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Szilvia Szabo
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary; Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Kata Juhasz
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Peter Kiraly
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Barbara Kocsis-Deak
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Beata Hargitai
- West Midlands Perinatal Pathology Centre, Cellular Pathology Department, Birmingham Women's and Children's NHS FT, Birmingham, United Kingdom
| | - Tibor Krenacs
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Petronella Hupuczi
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
| | - Offer Erez
- Maternity Department "D" Division of Obstetrics and Gynecology, Soroka University Medical Center, School of Medicine, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Zoltan Papp
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
| | - Ilona Kovalszky
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Nandor Gabor Than
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary; Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
50
|
Kaya B, Turhan U, Sezer S, Bestel A, Okumuş ZG, Dağ İ, Erel Ö. Maternal serum TXNDC5 levels and thiol/disulfide homeostasis in preeclamptic pregnancies. J Matern Fetal Neonatal Med 2018; 33:671-676. [PMID: 30522366 DOI: 10.1080/14767058.2018.1557140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Objective: To investigate thiol/disulfide homeostasis (TDH) and thioredoxin domain-containing 5 (TXNDC5) level in early and late-onset preeclampsia.Material and methods: In this cross-sectional study, 24 pregnant women with early-onset preeclampsia and 26 pregnant women with late-onset preeclampsia were compared with 30 pregnant women with no obstetric complications. The serum TXNDC5 levels and thiol/disulfide homeostasis were measured.Results: Serum TXNDC5 levels were significantly higher in the early-onset and late-onset preeclampsia groups compared with the control group (p < .05). Native thiol and total thiol levels were significantly lower in the early-onset and late-onset preeclampsia groups than control group. The disulfide levels were found as significantly high in early preeclamptic patients compared to control group (p < .05). The highest levels of TXNDC5 and the lowest levels of native thiol and total thiol were found in early-onset preeclampsia group. No significant difference was found between the patients with early onset and late onset preeclampsia regarding TXNDC5 levels and thiol/disulfide homeostasis (p > .05).Conclusion: Serum TXNDC5 levels were significantly higher in patients with early-onset and late-onset preeclampsia. The dynamic thiol/disulfide homeostasis was impaired in favor of the oxidized state in patients with preeclampsia.
Collapse
Affiliation(s)
- Başak Kaya
- Department of Maternal-Fetal Medicine, Health Sciences University, Antalya Education and Research Hospital, Antalya, Turkey
| | - Uğur Turhan
- Department of Maternal-Fetal Medicine, Health Sciences University, Kanuni Sultan Süleyman Education and Research Hospital, İstanbul, Turkey
| | - Salim Sezer
- Department of Maternal-Fetal Medicine, Health Sciences University, Kanuni Sultan Süleyman Education and Research Hospital, İstanbul, Turkey
| | - Ayşegül Bestel
- Department of Obstetrics and Gynecology, Health Sciences University, Kanuni Sultan Süleyman Education and Research Hospital, İstanbul, Turkey
| | - Zihniye Gonca Okumuş
- Department of Obstetrics and Gynecology, Health Sciences University, Kanuni Sultan Süleyman Education and Research Hospital, İstanbul, Turkey
| | - İsmail Dağ
- Department of Clinical Biochemistry, Eyüp State Hospital, İstanbul, Turkey
| | - Özcan Erel
- Department of Clinical Biochemistry, Yıldırım Beyazıt University, Ankara, Turkey
| |
Collapse
|