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
|
Alan M, Sorsa T, Meriç Kantar P, Raisanen IT, Gürlek Ö, Kanmaz B, Buduneli N. Active-Matrix Metalloproteinase-8, Myeloperoxidase in Relation With Periodontics, Preterm Birth. Oral Dis 2025; 31:1333-1341. [PMID: 39543825 DOI: 10.1111/odi.15202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/30/2024] [Accepted: 11/02/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE To investigate serum, placental levels of active-matrix metalloproteinase-8 (aMMP-8), myeloperoxidase (MPO) in preterm-birth with/without pre-eclampsia and term counterparts in relation with clinical periodontal parameters. METHODS Clinical periodontal measurements were recorded. Serum and placenta samples were collected during 173 full-term (FT), pre-term (PT) or pre-term complicated by pre-eclampsia (PTPE) deliveries. aMMP-8 levels were measured by IFMA. MPO levels in the serum and placenta samples were determined by ELISA. Data were tested using non-parametric tests. RESULTS PTBE group exhibited higher full-mouth probing depth and clinical attachment loss values than the other two groups (p < 0.05). Percentages of sites with plaque and bleeding on probing were lower in the PTBE group than in the other groups (p < 0.05). Serum aMMP-8 and MPO concentrations were higher in PTPE group than in the other groups (p < 0.05). Placenta aMMP-8 level was higher in the control group than in the PTPE group (p < 0.05). There was no significant difference between the groups in the placenta MPO levels (p > 0.05). CONCLUSIONS Within the limits of this cross-sectional study, it may be suggested that serum aMMP-8 and MPO concentrations together with placenta aMMP-8 levels may be associated with and reflect adverse pregnancy outcomes. Clinical periodontal findings did not reveal significant associations with these proteolytic and oxidative biomarkers.
Collapse
Affiliation(s)
- Murat Alan
- Department of Obstetrics and Gynecology, Izmir Tepecik Training and Research Hospital, Izmir, Turkey
| | - Timo Sorsa
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
- Department of Oral Diseases, Karolinska Institutet, Huddinge, Sweden
| | - Pınar Meriç Kantar
- Faculty of Dentistry, Department of Periodontology, Ege University, İzmir, Turkey
| | - Ismo T Raisanen
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Önder Gürlek
- Faculty of Dentistry, Department of Periodontology, Ege University, İzmir, Turkey
| | - Burcu Kanmaz
- Faculty of Dentistry, Department of Periodontology, İzmir Demokrasi University, İzmir, Turkey
| | - Nurcan Buduneli
- Faculty of Dentistry, Department of Periodontology, Ege University, İzmir, Turkey
| |
Collapse
|
3
|
Mishra JS, Bosse B, Hoppe KK, Malecki K, Hetzel SJ, Kumar S. Perfluoroalkyl substances (PFAS) exposure and preeclampsia risk: Impaired angiogenesis through suppression of VEGF signaling. Reprod Toxicol 2025; 132:108827. [PMID: 39732411 PMCID: PMC11890960 DOI: 10.1016/j.reprotox.2024.108827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/11/2024] [Accepted: 12/25/2024] [Indexed: 12/30/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are linked to preeclampsia (PE), a condition involving abnormal angiogenesis. Prior research on this association has been inconclusive. We investigated the relationship between maternal PFAS exposure and PE risk in Wisconsin. We also examined if PFAS disrupts angiogenesis and, if so, what mechanisms are involved. We conducted a case-control study with 40 PE cases and 40 controls. Maternal serum was analyzed for 38 different PFAS compounds using LC MS/MS. Functional in vitro experiments assessed PFOS effects on angiogenesis and mechanisms. Maternal serum samples from women with PE exhibited significantly higher PFOS and PFHPS concentrations than controls. After adjusting for confounders, each log-scale IQR increase in PFOS and PFHPS concentrations was associated with a 7.18-fold (95 % CI: 2.24, 23.0) and 5.40-fold (95 % CI: 1.81, 16.1) higher odds of PE, respectively. Furthermore, PFOS and PFHPS were positively associated with sFLT1 levels and the sFLT1/PLGF ratio. In vitro experiments revealed that PFOS exposure impaired HUVEC proliferation, migration, and tube formation, essential processes for angiogenesis. The membrane-based antibody array showed that PFOS decreased expression of multiple angiogenic proteins, including I-TAC, uPAR, VEGFR2, MMP-1, IL-1α, Angiopoietin-2, IL-1β, PECAM-1, TIE-2, and TIMP-2. The qPCR analysis demonstrated that PFOS decreased VEGFR2, the upstream target of VEGF, at the transcriptional level. In conclusion, elevated PFAS, especially PFOS and PFHPS, are linked to increased PE risk. PFOS may suppress angiogenesis via attenuated VEGFR2-mediated signaling, providing a molecular mechanism linking PFAS and PE pathogenesis.
Collapse
Affiliation(s)
- Jay S Mishra
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
| | - Bradley Bosse
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Kara K Hoppe
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Kristen Malecki
- Division of Environmental and Occupational Health Sciences, University of Illinois Chicago School of Public Health, Chicago, IL, USA
| | - Scott J Hetzel
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Sathish Kumar
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA; Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
4
|
Palma Dos Reis CR, O'Sullivan J, Ohuma EO, James T, Papageorghiou AT, Vatish M, Cerdeira AS. The ratio of soluble fms-like tyrosine kinase 1 to placental growth factor predicts time to delivery and mode of birth in patients with suspected preeclampsia: a secondary analysis of the INSPIRE trial. Am J Obstet Gynecol 2025; 232:317.e1-317.e17. [PMID: 38897339 DOI: 10.1016/j.ajog.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 05/23/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND The ratio of soluble fms-like tyrosine kinase 1 to placental growth factor is a useful biomarker for preeclampsia. Since it is a measure of placental dysfunction, it could also be a predictor of clinical deterioration and fetal tolerance to intrapartum stress. OBJECTIVE We tested the hypothesis that soluble fms-like tyrosine kinase 1 to placental growth factor ratio predicts time to delivery. Secondary objectives were to examine associations between the soluble fms-like tyrosine kinase 1 to placental growth factor ratio and mode of birth, fetal distress, need for labor induction, and birthweight z score. STUDY DESIGN Secondary analysis of the INSPIRE trial, a randomized interventional study on prediction of preeclampsia/eclampsia in which women with suspected preeclampsia were recruited and their blood soluble fms-like tyrosine kinase 1 to placental growth factor ratio was assessed. We stratified participants into 3 groups according to the ratio result: category 1 (soluble fms-like tyrosine kinase 1 to placental growth factor ≤38); category 2 (soluble fms-like tyrosine kinase 1 to placental growth factor >38 and <85); and category 3 (soluble fms-like tyrosine kinase 1 to placental growth factor ≥85). We modeled time from soluble fms-like tyrosine kinase 1 to placental growth factor determination to delivery using Kaplan-Meier curves and compared the 3 ratio categories adjusting for gestational age at soluble fms-like tyrosine kinase 1 to placental growth factor determination and trial arm with Cox regression. The association between ratio category and mode of delivery, induction of labor, and fetal distress was assessed using a multivariable logistic regression adjusting for gestational age at sampling and trial arm. The association between birthweight z score and soluble fms-like tyrosine kinase 1 to placental growth factor ratio was evaluated using multiple linear regression. Subgroup analysis was conducted in women with no preeclampsia and spontaneous onset of labor; women with preeclampsia; and participants in the nonreveal arm. RESULTS Higher ratio categories were associated with a shorter latency from soluble fms-like tyrosine kinase 1 to placental growth factor determination to delivery (37 vs 13 vs 10 days for ratios categories 1-3 respectively), hazards ratio for category 3 ratio of 5.64 (95% confidence interval 4.06-7.84, P<.001). A soluble fms-like tyrosine kinase 1 to placental growth factor ratio ≥85 had specificity of 92.7% (95% confidence interval 89.0%-95.1%) and sensitivity of 54.72% (95% confidence interval, 41.3-69.5) for prediction of preeclampsia indicated delivery within 2 weeks. A ratio category 3 was also associated with decreased odds of spontaneous vaginal delivery (Odds ratio [OR] 0.47, 95% confidence interval 0.25-0.89); an almost 6-fold increased risk of emergency cesarean section (OR 5.89, 95% confidence interval 3.05-11.21); and a 2-fold increased risk for intrapartum fetal distress requiring operative delivery or cesarean section (OR 3.04, 95% confidence interval 1.53-6.05) when compared to patients with ratios ≤38. Higher ratio categories were also associated with higher odds of induction of labor when compared to ratios category 1 (category 2, OR 2.20, 95% confidence interval 1.02-4.76; category 3, OR 6.0, 95% confidence interval 2.01-17.93); and lower median birthweight z score. Within subgroups of women a) without preeclampsia and with spontaneous onset of labor and b) women with preeclampsia, the log ratio was significantly higher in patients requiring intervention for fetal distress or failure to progress compared to those who delivered vaginaly without intervention. In the subset of women with no preeclampsia and spontaneous onset of labor, those who required intervention for fetal distress or failure to progress had a significantly higher log ratio than those who delivered vaginaly without needing intervention. CONCLUSION The soluble fms-like tyrosine kinase 1 to placental growth factor ratio might be helpful in risk stratification of patients who present with suspected preeclampsia regarding clinical deterioration, intrapartum fetal distress, and mode of birth (including the need for intervention in labor).
Collapse
Affiliation(s)
| | | | - Eric O Ohuma
- Maternal, Adolescent, Reproductive and Child Health (MARCH) Centre, London School of Hygiene and Tropical Medicine (LSHTM), London, UK
| | - Tim James
- Department of Clinical Biochemistry, Oxford University Hospitals NHS Foundation Trust, UK
| | | | - Manu Vatish
- Nuffield Department of Women's and Reproductive Health, University of Oxford, UK
| | - Ana Sofia Cerdeira
- Nuffield Department of Women's and Reproductive Health, University of Oxford, UK
| |
Collapse
|
5
|
Gehrs S, Jakab M, Gutjahr E, Gu Z, Weichenhan D, Mallm JP, Mogler C, Schlesner M, Plass C, Schlereth K, Augustin HG. The spatial zonation of the murine placental vasculature is specified by epigenetic mechanisms. Dev Cell 2025:S1534-5807(24)00776-7. [PMID: 39814015 DOI: 10.1016/j.devcel.2024.12.037] [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: 11/06/2023] [Revised: 08/08/2024] [Accepted: 12/18/2024] [Indexed: 01/18/2025]
Abstract
The labyrinthian fetoplacental capillary network is vital for proper nourishment of the developing embryo. Dysfunction of the maternal-fetal circulation is a primary cause of placental insufficiency. Here, we show that the spatial zonation of the murine placental labyrinth vasculature is controlled by flow-regulated epigenetic mechanisms. Spatiotemporal transcriptomic profiling identified a gradual change in the expression of epigenetic enzymes, including the de novo DNA methyltransferase 3a (DNMT3A). Loss of Dnmt3a resulted in DNA hypomethylation and perturbation of zonated placental gene expression. The resulting global DNA hypomethylation impaired the angiogenic capacity of endothelial cells. Global or endothelium-predominant deletion of Dnmt3a resulted in impaired placental vascularization and fetal growth retardation (FGR). Human placental endothelial gene expression profiling associated preeclampsia with reduced DNMT3A expression. Collectively, our study identified DMNT3A as critical methylome-regulator of placental endothelial gene expression and function with clinical implications for placental dysfunction, as it occurs during preeclampsia or FGR.
Collapse
Affiliation(s)
- Stephanie Gehrs
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany.
| | - Moritz Jakab
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Ewgenija Gutjahr
- Institute of Pathology, University Clinic Heidelberg, 69120 Heidelberg, Germany
| | - Zuguang Gu
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Dieter Weichenhan
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jan-Philipp Mallm
- Division of Chromatin Networks, German Cancer Research Center (DKFZ) and Bioquant, 69120 Heidelberg, Germany
| | - Carolin Mogler
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, 80333 Munich, Germany
| | - Matthias Schlesner
- Biomedical Informatics, Data Mining and Data Analytics, Faculty of Applied Computer Science and Medical Faculty, University of Augsburg, 86159 Augsburg, Germany
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Katharina Schlereth
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany.
| |
Collapse
|
6
|
Rutherford JN, George EK, Erickson EN. Placental biomarkers in second trimester maternal serum are associated with postpartum hemorrhage: a secondary analysis of the NuMoM2b dataset. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.12.30.24319779. [PMID: 39802772 PMCID: PMC11722460 DOI: 10.1101/2024.12.30.24319779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Postpartum hemorrhage (PPH) is the leading cause of maternal mortality worldwide, which is often attributed to retained placenta (RP) after delivery. There are no biomarkers currently used to predict a risk of developing RP/PPH prior to labor. The objective of this study was to determine relationships between placental biomarkers measured in the first and second trimesters and proxy measures of postpartum blood loss relative to preeclampsia status in the Nulliparous Pregnancy Outcomes Study: Monitoring Mothers-to-Be (nuMoM2b) dataset. 2,192 participants had placental analytes drawn during the first and second trimesters (9-13 and 16-22 weeks gestation, respectively); the outcome was a composite of retained placenta and/or PPH requiring blood transfusion (RP/PPH). Using Kruskal-Wallis tests, median differences in levels of soluble fms-like tyrosine kinase-1 (sFlt-1), placental growth factor (PlGF), sFlt-1/PlGF ratio, soluble endoglin (sEng), beta subunit of human chorionic gonadotropin (β-hCG), inhibin A (INHA), and pregnancy-associated protein-A (PAPP-A) were assessed between women with (n=67) and without (n=2125) RP/PPH overall and stratified by preeclampsia status. Women with RP/PPH had significantly higher median levels of sEng, β-hCG, INHA, PAPP-A in the second trimester and sFlt-1was higher in both first and second trimesters, which was observed again when stratifying by preeclampsia status. Our findings indicate that biomarkers associated with angiogenesis, particularly when measured in the second trimester, are important targets for further study of RP and/or PPH pathophysiology and potential risk screening development.
Collapse
Affiliation(s)
- Julienne N. Rutherford
- University of Arizona, College of Nursing, Division of Nursing and Health Sciences, Tucson, Arizona
| | - Erin K. George
- University of Arizona, College of Nursing, Division of Nursing and Health Sciences, Tucson, Arizona
| | - Elise N. Erickson
- University of Arizona, College of Nursing, Division of Nursing and Health Sciences, Tucson, Arizona
| |
Collapse
|
7
|
Hashmat A, Ya J, Kadir R, Alwjwaj M, Bayraktutan U. Hyperglycaemia perturbs blood-brain barrier integrity through its effects on endothelial cell characteristics and function. Tissue Barriers 2025; 13:2350821. [PMID: 38712515 PMCID: PMC11970753 DOI: 10.1080/21688370.2024.2350821] [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: 02/21/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/08/2024] Open
Abstract
Breakdown of blood-brain barrier (BBB) represents a key pathology in hyperglycemia-mediated cerebrovascular damage after an ischemic stroke. As changes in the level and nature of vasoactive agents released by endothelial cells (ECs) may contribute to BBB dysfunction, this study first explored the specific impact of hyperglycemia on EC characteristics and secretome. It then assessed whether secretome obtained from ECs subjected to normoglycaemia or hyperglycemia might regulate pericytic cytokine profile differently. Using a triple cell culture model of human BBB, composed of brain microvascular EC (BMEC), astrocytes and pericytes, this study showed that exposure to hyperglycemia (25 mM D-glucose) for 72 h impaired the BBB integrity and function as evidenced by decreases in transendothelial electrical resistance and increases in paracellular flux of sodium fluorescein. Dissolution of zonula occludens-1, a tight junction protein, and appearance of stress fibers appeared to play a key role in this pathology. Despite elevations in angiogenin, endothelin-1, interleukin-8 and basic fibroblast growth factor levels and a decrease in placental growth factor levels in BMEC subjected to hyperglycemia vs normoglycaemia (5.5 mM D-glucose), tubulogenic capacity of BMECs remained similar in both settings. Similarly, pericytes subjected to secretome obtained from hyperglycemic BMEC released higher quantities of macrophage migration inhibitory factor and serpin and lower quantities of monocyte chemoattractant protein-1, intercellular adhesion molecule, interleukin-6 and interleukin-8. Taken together these findings indicate the complexity of the mechanisms leading to BBB disruption in hyperglycemic settings and emphasize the importance of endothelial cell-pericyte axis in the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Arshad Hashmat
- Academic Unit of Mental Health and Clinical Neurosciences, School of Medicine, The University of Nottingham, Nottingham, UK
| | - Jingyuan Ya
- Academic Unit of Mental Health and Clinical Neurosciences, School of Medicine, The University of Nottingham, Nottingham, UK
| | - Rais Kadir
- Academic Unit of Mental Health and Clinical Neurosciences, School of Medicine, The University of Nottingham, Nottingham, UK
| | - Mansour Alwjwaj
- Academic Unit of Mental Health and Clinical Neurosciences, School of Medicine, The University of Nottingham, Nottingham, UK
| | - Ulvi Bayraktutan
- Academic Unit of Mental Health and Clinical Neurosciences, School of Medicine, The University of Nottingham, Nottingham, UK
| |
Collapse
|
8
|
Prasetyorini N, Erwan NE, Sardjono TW, Nurseta T, Utomo RP, Nugraha RYB, Cahayani WA, Rukmigarsari E, Arinugraha LN, Fitri LE. HIF-1α regulated pathomechanism of low birth weight through angiogenesis factors in placental Plasmodium vivax infection. F1000Res 2024; 11:131. [PMID: 38884107 PMCID: PMC11179053 DOI: 10.12688/f1000research.73820.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/03/2024] [Indexed: 06/18/2024] Open
Abstract
Background Malaria in pregnancy leads to placental malaria. The primary pathogenesis of the complex fetal implications in placental malaria is tissue hypoxia due to sequestrations of Plasmodium falciparum-infected erythrocytes in the placenta. However, the pathomechanism of placental Plasmodium vivax infection has not been thoroughly investigated. Hypoxia-inducible factor-1α (HIF-1α) is a key transcriptional mediator of the response to hypoxic conditions, which interacts with the change and imbalances of many chemical mediators, including angiogenic factors, leading to fetal growth abnormality. Methods This study was conducted cross-sectionally in Maumere, Sikka Regency, East Nusa Tenggara Province, previously known as one of the malaria endemic areas with a high incidence of low birth weight (LBW) cases. This study collected peripheral and umbilical blood samples and placental tissues from mothers who delivered their babies with LBW at the TC Hiller Regional Hospital. All of the blood samples were examined for parasites by microscopic and PCR techniques, while the plasma levels of VEGF, PlGF, VEGFR-1, VEGFR-2, and HIF-1α were determined using ELISA. The sequestration of infected erythrocytes and hemozoin was determined from placental histological slides, and the expression of placenta angiogenic factors was observed using the immunofluorescent technique. Results In this study, 33 cases had complete data to be analyzed. Of them, 19 samples were diagnosed as vivax malaria and none of falciparum malaria. There were significant differences in Δ 10th percentile growth curve of baby's body weights and also all angiogenic factors in placental tissues {VEGF, PlGF, and VEGFR-1, VEGFR-2, and HIF-1α} between those infected and not infected cases (p<0.05), but not for VEGF and VEGFR-2 in the plasma. Conclusion This study indicated that Plasmodium vivax sequestration may promote LBW through alterations and imbalances in angiogenic factors led by HIF-1α.
Collapse
Affiliation(s)
- Nugrahanti Prasetyorini
- Doctoral Program in Medical Science, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Malaria Research Group, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Department of Obstetrics & Gynecology, Faculty of Medicine Universitas Brawijaya/dr Saiful Anwar Hospital, Malang, Indonesia
| | - Nabila Erina Erwan
- Malaria Research Group, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Master Program in Biomedical Science, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Teguh Wahju Sardjono
- Malaria Research Group, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Department of Parasitology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Tatit Nurseta
- Department of Obstetrics & Gynecology, Faculty of Medicine Universitas Brawijaya/dr Saiful Anwar Hospital, Malang, Indonesia
| | - Rudi Priyo Utomo
- Department of Obstetrics & Gynecology, dr T.C. Hillers Regional Hospital, Maumere, Sikka Regency, NTT, Indonesia
| | - Rivo Yudhinata Brian Nugraha
- Malaria Research Group, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Department of Parasitology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Wike Astrid Cahayani
- Malaria Research Group, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Department of Anatomy and Histology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Ettie Rukmigarsari
- Mathematics Education Study Program, Faculty of Teacher Training and Education, University of Islam Malang, Malang, Indonesia
| | | | - Loeki Enggar Fitri
- Malaria Research Group, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Department of Parasitology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| |
Collapse
|
9
|
Tamil Barathi P, Mohanapriya A. Pre-eclampsia: Re-visiting pathophysiology, role of immune cells, biomarker identification and recent advances in its management. J Reprod Immunol 2024; 163:104236. [PMID: 38555746 DOI: 10.1016/j.jri.2024.104236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/21/2024] [Accepted: 03/07/2024] [Indexed: 04/02/2024]
Abstract
Pre-eclampsia (PE) is a hypertension condition that occurs exclusively during pregnancy and has the potential to impact nearly all organ systems. It is estimated to complicate approximately 2-8% of pregnancies worldwide. PE is a prominent medical disorder that poses a significant risk to pregnant mothers and their infants. This review commences by giving the most up-to- date concepts about the pathophysiology of PE. The condition involves atypical infiltration of trophoblast cells into the spiral arteries of the decidua and myometrium, resulting in an insufficient establishment of proper blood flow between the uterus and placenta. The aberrant activation of natural killer (NK) cells in both the peripheral blood and the decidua has been identified as one of the contributing factors to the development of PE. The strong evidence for the genetic etiology of PE is provided by the association between maternal killer cell immunoglobulin-like receptor (KIR) and Human Leukocyte Antigen (HLA-C) in trophoblast cells. Recent observations provide evidence that changes in the expression of anti-angiogenic factors in the placenta are the underlying cause of the clinical symptoms associated with the condition. This review also provides a comprehensive overview of the latest advancements in understanding the underlying causes of PE. It specifically highlights the emergence of new diagnostic biomarkers and their potential implications for therapeutic interventions in managing this medical condition.
Collapse
Affiliation(s)
- Palanisamy Tamil Barathi
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India.
| | - Arumugam Mohanapriya
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India.
| |
Collapse
|
10
|
Tomescu LC, Sas I, Sarb S, Cimpean AM. Evaluation of Vasculogenic Factors in the Developing Embryo at Weeks Five and Seven With a Special Focus on CD133 and TIE2 Markers. Cureus 2024; 16:e60353. [PMID: 38756714 PMCID: PMC11096276 DOI: 10.7759/cureus.60353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 05/18/2024] Open
Abstract
Background Human embryo vasculogenesis (blood vessel development starting from endothelial precursors) includes the ability of mesenchymal cells and pluripotent stem cells to differentiate into endothelial cells. Quantification of endothelial progenitor cells is difficult to assess during the early steps of human embryo development due to several factors, especially due to the paucity of human embryo tissue which is usually discarded after early-stage pregnancy abortive methods. CD133 (Promimin-1) is a general marker of progenitor cells, but combined with other endothelial markers such as CD34, it may identify endothelial progenitor cells during embryonic development. CD34 immunohistochemistry was previously performed by our team to identify human embryo capillaries and comparatively assess microvessel density between different human embryonic tissues. TIE2 is an angiopoietin receptor strongly involved in the newly formed blood vessel maturation due to its expression in some mesenchymal precursors for future pericytes. CD34 assesses the presence of endothelial cells but its single use does not evaluate the endothelial progenitor state as CD133 may do nor vessel maturation as TIE2 may do. Data about the dynamics of CD133/TIE2 expression in the early stages of human embryo development are scarce. Hence, in this study, we aimed to comparatively assess the dynamic of CD133+ endothelial precursors and TIE2 expression on five and seven-week-old human embryonic tissues with a special emphasis on their expression on embryonic vascular beds. Methodology CD133 and TIE2 immunohistochemistry was performed on five and seven-week-old human embryonic tissues followed by their quantification using the Qu Path digital image analysis (DIA) automated method. Results CD133 and TIE2 showed divergent patterns of expression during the initial phases of human embryonic development, specifically in the vascular endothelium of tiny capillaries. The expression of CD133 in endothelial cells lining the perfused lumen gradually decreased from five to seven-week-old embryos. It remained expressed with greater intensity in cells located at the tip of the vascular bud that emerged into pre-existing capillaries. TIE2 was much more specific than CD133, being restricted to the level of the vascular endothelium; therefore, it was easier to quantify using digital image analysis. The endothelium of the embryonic aorta was an exception to the divergent expression, as CD133 and TIE2 were consistently co-expressed in the seven-week-old embryo. The Qu Path DIA assessment increased the accuracy of CD133 and TIE2 evaluation, being the first time they were quantified by using automated software and not manually. Conclusions High heterogeneity of CD133 and TIE2 was observed between five and seven-week-old embryonic tissues as well as between different embryonic regions from the same gestational age. The unique finding of CD133/TIE2 co-expression persistence inside aortic endothelium needs further studies to elucidate the role of this co-expression.
Collapse
Affiliation(s)
| | - Ioan Sas
- Obstetrics and Gynecology, Victor Babeş University of Medicine and Pharmacy, Timișoara, ROU
| | - Simona Sarb
- Microscopic Morphology/Histology, Victor Babeş University of Medicine and Pharmacy, Timișoara, ROU
| | - Anca Maria Cimpean
- Microscopic Morphology/Histology, Victor Babeş University of Medicine and Pharmacy, Timișoara, ROU
| |
Collapse
|
11
|
Theerathananon W, Watanapa WB, Wataganara T, Pratumvinit B, Rahman S. Preeclamptic serum and soluble fms-like tyrosine kinase-1 suppress endothelial inward rectifier potassium currents. Placenta 2024; 146:101-109. [PMID: 38241839 DOI: 10.1016/j.placenta.2024.01.002] [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: 08/09/2023] [Revised: 11/01/2023] [Accepted: 01/03/2024] [Indexed: 01/21/2024]
Abstract
INTRODUCTION Inward rectifier K+ (Kir) channel, a major factor determining endothelial membrane potential, regulates Ca2+ influx and vasodilator release, which is impaired in preeclamptic blood vessels. Previously, human umbilical vein endothelial cell (HUVEC) Kir currents were shown to decrease after incubating in preeclamptic plasma. We aimed to demonstrate whether sFlt-1, which is high in preeclamptic blood, could inhibit Kir channel function and expression. METHODS HUVECs were cultured in regular medium, regular medium with added sFlt-1, or serum from preeclampsia patients or normal pregnant women (Control, sFlt-1, PE, or NP, respectively). Using whole-cell patch clamp technique, we identified Kir currents with the Kir blocker 2 mM BaCl2 and compared the currents among groups. The expression of Kir 2.1 and 2.2 channels were determined using immunofluorescent staining. RESULTS sFlt-1 and PE groups exhibited similar Kir currents, while NP group possessed significantly larger currents, similar to Control group currents. Moreover, sFlt-1 and sFlt-1/PlGF ratio showed strong negative correlation with Kir currents (r = -0.71 and -0.70, respectively; P < 0.05). There were no significant differences in mean fluorescence intensity representing Kir 2.1 and 2.2 channels expression in all four groups. DISCUSSION This is the first report to demonstrate sFlt-1 inhibition against Kir currents, which could lead to maternal endothelial dysfunction and hypertension seen in preeclampsia. However, channel expression was unaffected by sFlt-1 incubation, suggesting dysfunctions of channel or other processes (e.g., membrane translocation). The present data could pave the way for novel therapies targeting sFlt-1 or Kir to alleviate hypertension in preeclampsia.
Collapse
Affiliation(s)
- Wuttinan Theerathananon
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkok, 10700, Thailand.
| | - Wattana B Watanapa
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkok, 10700, Thailand.
| | - Tuangsit Wataganara
- Department of Obstetrics and Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkok, 10700, Thailand.
| | - Busadee Pratumvinit
- Department of Clinical Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkok, 10700, Thailand.
| | - Suraiya Rahman
- Department of Obstetrics and Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkok, 10700, Thailand.
| |
Collapse
|
12
|
Nacar MC, Yigit S, Keskin A, Kirici P, Karacor T. VEGF I/D variant and preeclampsia risk in Turkish women: a case-control study. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2023; 43:585-595. [PMID: 37991450 DOI: 10.1080/15257770.2023.2283621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
Preeclampsia (PE) is a systemic vascular disorder, is caused by an imbalance of pro- and anti-angiogenic factors that directly affect endothelial function. Vascular endothelial growth factor A (called VGF), a pro-angiogenic factor associated with endothelial dysfunction, plays an important role in the pathophysiology of PE. Therefore, we investigated the relationship between -2549 insertion/deletion (I/D) variant in the VEGF promoter region and PE in pregnant women in Turkey. A total of 100 patients diagnosed with PE and 118 healthy pregnants were recruited. To genotype the VEGF I/D variant, the PCR method was used. The results of analyses were evaluated for statistical significance. The weight of the PE group was found to be higher before and after pregnancy than the control group (p = 0.009, p = 0.012, respectively). The birth weight, and Apgar score (1 min and 5 min) of the PE group was lower than that of the control group (p= <0.001, p= <0.001, p= <0.001, respectively). The mean 24-h urine protein, ALT and AST levels in the PE group were higher than the control group (p= <0.001, p= <0.001, p= <0.001, respectively). There was no significant difference between the patients and the controls in terms of VEGF I/D genotype and allele distribution. There was no deviation from HWA for VEGF I/D variant in patient and control groups. In the patients carrying D/D genotype and the D allele had low gestational week and birth weight. Knowing the risk factors for PE is very important for its prevention and treatment. In conclusion, for the first time, our results supported that the VEGF I/D variant is not a risk factor for the development of PE in a group of Turkish populations. But VEGF I/D variant D/D genotype associated with low gestational week and birth weight while I/D genotype seems to be protective from high systolic blood pressure.
Collapse
Affiliation(s)
- Mehmet Can Nacar
- Department of Obstetrics and Gynecology, Adiyaman University School of Medicine, Adiyaman, Turkiye
| | - Serbulent Yigit
- Department of Genetics, Faculty of Veterinary, Ondokuz Mayis University, Samsun, Turkiye
- Department of Medical Biology, Graduate Institute, Ondokuz Mayis University, Samsun, Turkiye
| | - Adem Keskin
- Department of Biochemistry Institute of Health Sciences, Adnan Menderes University, Aydin, Turkiye
| | - Pinar Kirici
- Department of Obstetrics and Gynecology, Adiyaman University School of Medicine, Adiyaman, Turkiye
| | - Talip Karacor
- Department of Obstetrics and Gynecology, Adiyaman University School of Medicine, Adiyaman, Turkiye
| |
Collapse
|
13
|
Covarrubias A, Aguilera-Olguín M, Carrasco-Wong I, Pardo F, Díaz-Astudillo P, Martín SS. Feto-placental Unit: From Development to Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:1-29. [PMID: 37466767 DOI: 10.1007/978-3-031-32554-0_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
The placenta is an intriguing organ that allows us to survive intrauterine life. This essential organ connects both mother and fetus and plays a crucial role in maternal and fetal well-being. This chapter presents an overview of the morphological and functional aspects of human placental development. First, we describe early human placental development and the characterization of the cell types found in the human placenta. Second, the human placenta from the second trimester to the term of gestation is reviewed, focusing on the morphology and specific pathologies that affect the placenta. Finally, we focus on the placenta's primary functions, such as oxygen and nutrient transport, and their importance for placental development.
Collapse
Affiliation(s)
- Ambart Covarrubias
- Health Sciences Faculty, Universidad San Sebastián, Concepción, Chile
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Macarena Aguilera-Olguín
- Biomedical Research Centre, School of Medicine, Universidad de Valparaíso, Viña del Mar, Chile
- Cellular Signalling and Differentiation Laboratory (CSDL), Medicine and Science Faculty, Universidad San Sebastián, Santiago, Chile
| | - Ivo Carrasco-Wong
- Cellular Signalling and Differentiation Laboratory (CSDL), School of Medical Technology, Medicine and Science Faculty, Universidad San Sebastián, Santiago, Chile
| | - Fabián Pardo
- Metabolic Diseases Research Laboratory, Interdisciplinary Centre of Territorial Health Research (CIISTe), Biomedical Research Center (CIB), San Felipe Campus, School of Medicine, Faculty of Medicine, Universidad de Valparaíso, San Felipe, Chile
| | - Pamela Díaz-Astudillo
- Biomedical Research Centre, School of Medicine, Universidad de Valparaíso, Viña del Mar, Chile
| | - Sebastián San Martín
- Biomedical Research Centre, School of Medicine, Universidad de Valparaíso, Viña del Mar, Chile.
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile.
| |
Collapse
|
14
|
Wang LQ, Fernandez-Boyano I, Robinson WP. Genetic variation in placental insufficiency: What have we learned over time? Front Cell Dev Biol 2022; 10:1038358. [PMID: 36313546 PMCID: PMC9613937 DOI: 10.3389/fcell.2022.1038358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/03/2022] [Indexed: 11/28/2022] Open
Abstract
Genetic variation shapes placental development and function, which has long been known to impact fetal growth and pregnancy outcomes such as miscarriage or maternal pre-eclampsia. Early epidemiology studies provided evidence of a strong heritable component to these conditions with both maternal and fetal-placental genetic factors contributing. Subsequently, cytogenetic studies of the placenta and the advent of prenatal diagnosis to detect chromosomal abnormalities provided direct evidence of the importance of spontaneously arising genetic variation in the placenta, such as trisomy and uniparental disomy, drawing inferences that remain relevant to this day. Candidate gene approaches highlighted the role of genetic variation in genes influencing immune interactions at the maternal-fetal interface and angiogenic factors. More recently, the emergence of molecular techniques and in particular high-throughput technologies such as Single-Nucleotide Polymorphism (SNP) arrays, has facilitated the discovery of copy number variation and study of SNP associations with conditions related to placental insufficiency. This review integrates past and more recent knowledge to provide important insights into the role of placental function on fetal and perinatal health, as well as into the mechanisms leading to genetic variation during development.
Collapse
Affiliation(s)
- Li Qing Wang
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Icíar Fernandez-Boyano
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Wendy P. Robinson
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
15
|
Hebeda CB, Savioli AC, Scharf P, de Paula-Silva M, Gil CD, Farsky SHP, Sandri S. Neutrophil depletion in the pre-implantation phase impairs pregnancy index, placenta and fetus development. Front Immunol 2022; 13:969336. [PMID: 36248911 PMCID: PMC9558710 DOI: 10.3389/fimmu.2022.969336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Maternal neutrophils cells are players in gestational tolerance and fetus delivery. Nonetheless, their actions in each phase of the pregnancy are unknown. We here investigated the role of maternal neutrophil depletion before the blastocyst implantation phase and outcomes in the pregnancy index, placenta, and fetus development. Neutrophils were pharmacologically depleted by i.p. injection of anti-Gr1 (anti-neutrophils; 200 µg) 24 hours after plug visualization in allogeneic-mated C57BL/6/BALB/c mice. Depletion of peripheral neutrophils lasted until 48 hours after anti-Gr1 injection (gestational day 1.5-3.5). On gestational day 5.5, neutrophil depletion impaired the blastocyst implantation, as 50% of pregnant mice presented reduced implantation sites. On gestational day 18.5, neutrophil depletion reduced the pregnancy rate and index, altered the placenta disposition in the uterine horns, and modified the structure of the placenta, detected by reduced junctional zone, associated with decreased numbers of giant trophoblast cells, spongiotrophoblast. Reduced number of placenta cells labeled for vascular endothelial growth factor (VEGF), platelet-endothelial cell adhesion molecule (PECAM-1), and intercellular cell adhesion molecule (ICAM-1), important markers of angiogenesis and adhesiveness, were detected in neutrophil depleted mice. Furthermore, neutrophil depletion promoted a higher frequency of monocytes, natural killers, and T regulatory cells, and lower frequency of cytotoxic T cells in the blood, and abnormal development of offspring. Associated data obtained herein highlight the pivotal role of neutrophils actions in the early stages of pregnancy, and address further investigations on the imbricating signaling evoked by neutrophils in the trophoblastic interaction with uterine epithelium.
Collapse
Affiliation(s)
- Cristina Bichels Hebeda
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, São Paulo, Brazil
- Núcleo de Pesquisa em Ciências Médicas, Fundação Universidade para o Desenvolvimento do Alto Vale do Itajaí – UNIDAVI, Rio do Sul, SC, Brazil
| | - Anna Carolina Savioli
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, São Paulo, Brazil
| | - Pablo Scharf
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, São Paulo, Brazil
| | - Marina de Paula-Silva
- Center for Stem Cells and Regenerative Medicine, King’s College London, London, United Kingdom
| | - Cristiane Damas Gil
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Sandra Helena Poliselli Farsky
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, São Paulo, Brazil
| | - Silvana Sandri
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, São Paulo, Brazil
- *Correspondence: Silvana Sandri,
| |
Collapse
|
16
|
Tiwari P, Gupta MM, Jain SL. Placental findings in singleton stillbirths: a case-control study from a tertiary-care center in India. J Perinat Med 2022; 50:753-762. [PMID: 34587673 DOI: 10.1515/jpm-2021-0179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/11/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Placental examination in a case of stillbirth can provide insight into causative/associated factors with fetal demise. The aim of this study was to compare placental and umbilical cord pathologies in singleton stillbirth and livebirth placentas, and to find prevalence of various associated maternal and fetal clinical factors. METHODS This case-control study was conducted at a tertiary-care center in India over a period of 20 months. About 250 women who delivered stillborn fetus ≥28 weeks' gestation and 250 maternal-age-matched controls were recruited. Sociodemographic and clinical details were noted and placental gross and microscopic examination was done. Placental findings were compared between stillbirth and livebirth (overall), preterm stillbirth and preterm livebirth as well as term stillbirth and term livebirth in six categories - placenta gross, cord gross, membranes gross, maternal vascular malperfusion, fetal vascular malperfusion and inflammatory response. Prevalence of 11 maternal and fetal factors were studied in all categories of placental findings in both livebirth and stillbirth. RESULTS Placental findings in all six categories were significantly associated with stillbirths (p<0.05). The placental findings associated with stillbirth with highest odds included placental hypoplasia (OR 9.77, 95% CI 5.46-17.46), necrotizing chorioamnionitis (OR 9.30, 95% CI 1.17-73.96) and avascular villi (OR 8.45, 95% CI 3.53-20.25). More than half of the women with stillbirths had medical disorders (n=130, 52.0%) and the most prevalent was hypertensive disorder (n=45, 18.0%). CONCLUSIONS Changes in placenta are associated with development of stillbirth. Therefore, antenatal investigations to identify placental dysfunction should be investigated to determine whether these reduce stillbirth. Also, placental examination in a case of stillbirth can detect/diagnose many maternal/fetal conditions and thereby can help in preventing future stillbirths.
Collapse
Affiliation(s)
- Purnima Tiwari
- Department of Obstetrics & Gynaecology, All India Institute of Medical Sciences Nagpur, Nagpur, India
| | - Madhavi M Gupta
- Department of Obstetrics & Gynaecology, Maulana Azad Medical College and associated Lok Nayak Hospital, New Delhi, India
| | - Shyama Lata Jain
- Department of Pathology, Maulana Azad Medical College, New Delhi, India
| |
Collapse
|
17
|
Reactive Oxygen Species are Essential for Placental Angiogenesis During Early Gestation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4290922. [PMID: 35693704 PMCID: PMC9177322 DOI: 10.1155/2022/4290922] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/07/2022] [Indexed: 12/30/2022]
Abstract
Background Preeclampsia (PE) is associated with insufficient placental perfusion attributed to maldevelopment of the placental vasculature. Reactive oxygen species (ROS) are implicated in angiogenesis, but their regulatory effects and mechanisms in placental vascular development remain unclear. Methods Placental oxidative stress was determined throughout gestation by measuring 4-hydroxynonenal (4HNE) and malondialdehyde (MDA). The antioxidant MitoQ was administered to pregnant mice from GDs 7.5 to 11.5; placental morphology and angiogenesis pathways were examined on GDs 11.5 and 18.5. Moreover, we established a mouse mFlt-1-induced PE model and assessed blood pressure, urine protein levels, and placental vascular development on GDs 11.5 and 18.5. Human umbilical vein endothelial cells (HUVECs) were treated with various H2O2 concentrations to evaluate cell viability, intracellular ROS levels, and tube formation capability. MitoQ, an AKT inhibitor and an ERK1/2 inhibitor were applied to validate the ROS-mediated mechanism regulating placental angiogenesis. Results First-trimester placentas presented significantly higher MDA and 4HNE levels. MitoQ significantly reduced the blood vessel density and angiogenesis pathway activity in the placenta on GDs 11.5 and 18.5. Serum sFlt-1 levels were elevated, and we observed poor placental angiogenesis and PE-like symptoms in cases with mFlt-1 overexpression. Moderate H2O2 treatment promoted HUVEC proliferation and angiogenesis, whereas these improvements were abolished by MitoQ, AKT inhibitor, or ERK1/2 inhibitor treatment. Conclusions Moderate ROS levels are essential for placental angiogenesis; diminishing ROS with potent antioxidants during placentation decreases placental angiogenesis and increases PE risk. Therefore, antioxidant therapy should be considered carefully for normal pregnant women during early gestation.
Collapse
|
18
|
Zhang N, Schumacher A, Fink B, Bauer M, Zenclussen AC, Meyer N. Insights into Early-Pregnancy Mechanisms: Mast Cells and Chymase CMA1 Shape the Phenotype and Modulate the Functionality of Human Trophoblast Cells, Vascular Smooth-Muscle Cells and Endothelial Cells. Cells 2022; 11:cells11071158. [PMID: 35406722 PMCID: PMC8997408 DOI: 10.3390/cells11071158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 01/27/2023] Open
Abstract
Spiral-artery (SA) remodeling is a fundamental process during pregnancy that involves the action of cells of the initial vessel, such as vascular smooth-muscle cells (VSMCs) and endothelial cells, but also maternal immune cells and fetal extravillous trophoblast cells (EVTs). Mast cells (MCs), and specifically chymase-expressing cells, have been identified as key to a sufficient SA-remodeling process in vivo. However, the mechanisms are still unclear. The purpose of this study is to evaluate the effects of the MC line HMC-1 and recombinant human chymase (rhuCMA1) on human primary uterine vascular smooth-muscle cells (HUtSMCs), a human trophoblast cell line (HTR8/SV-neo), and human umbilical-vein endothelial cells (HUVEC) in vitro. Both HMC-1 and rhuCMA1 stimulated migration, proliferation, and changed protein expression in HUtSMCs. HMC-1 increased proliferation, migration, and changed gene expression of HTR8/SVneo cells, while rhuCMA treatment led to increased migration and decreased expression of tissue inhibitors of matrix metalloproteinases. Additionally, rhuCMA1 enhanced endothelial-cell-tube formation. Collectively, we identified possible mechanisms by which MCs/rhuCMA1 promote SA remodeling. Our findings are relevant to the understanding of this crucial step in pregnancy and thus of the dysregulated pathways that can lead to pregnancy complications such as fetal growth restriction and preeclampsia.
Collapse
Affiliation(s)
- Ningjuan Zhang
- Department of Environmental Immunology, UFZ-Helmholtz Centre for Environmental Research Leipzig-Halle, 04318 Leipzig, Germany; (N.Z.); (A.S.); (B.F.); (M.B.); (A.C.Z.)
- Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, University Leipzig, 04103 Leipzig, Germany
| | - Anne Schumacher
- Department of Environmental Immunology, UFZ-Helmholtz Centre for Environmental Research Leipzig-Halle, 04318 Leipzig, Germany; (N.Z.); (A.S.); (B.F.); (M.B.); (A.C.Z.)
- Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, University Leipzig, 04103 Leipzig, Germany
| | - Beate Fink
- Department of Environmental Immunology, UFZ-Helmholtz Centre for Environmental Research Leipzig-Halle, 04318 Leipzig, Germany; (N.Z.); (A.S.); (B.F.); (M.B.); (A.C.Z.)
| | - Mario Bauer
- Department of Environmental Immunology, UFZ-Helmholtz Centre for Environmental Research Leipzig-Halle, 04318 Leipzig, Germany; (N.Z.); (A.S.); (B.F.); (M.B.); (A.C.Z.)
| | - Ana Claudia Zenclussen
- Department of Environmental Immunology, UFZ-Helmholtz Centre for Environmental Research Leipzig-Halle, 04318 Leipzig, Germany; (N.Z.); (A.S.); (B.F.); (M.B.); (A.C.Z.)
- Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, University Leipzig, 04103 Leipzig, Germany
| | - Nicole Meyer
- Department of Environmental Immunology, UFZ-Helmholtz Centre for Environmental Research Leipzig-Halle, 04318 Leipzig, Germany; (N.Z.); (A.S.); (B.F.); (M.B.); (A.C.Z.)
- Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, University Leipzig, 04103 Leipzig, Germany
- Correspondence: ; Tel.: +49-341-235-1542
| |
Collapse
|
19
|
Li W, Liu S, Li Y, Wang W, Dong Y, Qi L, Ren J, Zhang J. Association between serum placental growth factor and vascular endothelial function in hyprtensive disorders complicating pregnancy. Exp Biol Med (Maywood) 2021; 247:480-487. [PMID: 34904900 DOI: 10.1177/15353702211063833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Hypertensive disorders complicating pregnancy (HDCP) is a systemic disease among pregnant women. Therefore, the prevention and prediction of hypertension during pregnancy are critical. This study aimed to clarify whether the vascular endothelial function of women with gestational hypertension was linked to placental growth factor. A total of 200 pregnant women were enrolled in our study and subsequently divided into two groups: the HDCP group and the normal pregnancy controls. The levels of serum placental growth factor, as well as plasma endothelin-1 and nitric oxide, between the two groups were measured. In addition, the endothelial function indexes, including pressure-strain elasticity coefficient (EP), the common carotid stiffness index (β), arterial compliance, single-point pulsed-wave velocity, and augment index (AI) of bilateral common carotid arteries, were compared between the HDCP and control groups using the echo tracking technique. In our study, the level of placental growth factor in the HDCP group was significantly lower than the control group. Furthermore, our results clarified that endothelin-1 increased while nitric oxide decreased in the HDCP group compared with the control group. On the other hand, we found that EP, β, pulsed-wave velocity and augment index values were significantly higher in the HDCP group than in the control group (P < 0.001). However, the value of arterial compliance was significantly decreased in patients of the HDCP group compared with the control group (P < 0.001). In conclusion, the association between serum placental growth factor and vascular endothelial function in HDCP could serve as a more accurate predictive factor of pregnant hypertension.
Collapse
Affiliation(s)
- Weiling Li
- Department of General Internal Medicine, Shijiazhuang Fourth Hospital, Shijiazhuang, Hebei Province 050031, China
| | - Shasha Liu
- Department of General Internal Medicine, Shijiazhuang Fourth Hospital, Shijiazhuang, Hebei Province 050031, China
| | - Yuan Li
- Department of General Internal Medicine, Shijiazhuang Fourth Hospital, Shijiazhuang, Hebei Province 050031, China
| | - Weijing Wang
- Department of Diagnostic Ultrasound, Shijiazhuang Fourth Hospital, Shijiazhuang, Hebei Province 050031, China
| | - Yiming Dong
- Department of General Internal Medicine, Shijiazhuang Fourth Hospital, Shijiazhuang, Hebei Province 050031, China
| | - Lin Qi
- Department of General Internal Medicine, Shijiazhuang Fourth Hospital, Shijiazhuang, Hebei Province 050031, China
| | - Jie Ren
- Department of Obstetrics, Shijiazhuang Fourth Hospital, Shijiazhuang, Hebei Province 050031, China
| | - Jiefang Zhang
- Department of General Internal Medicine, Shijiazhuang Fourth Hospital, Shijiazhuang, Hebei Province 050031, China
| |
Collapse
|
20
|
Munchel S, Rohrback S, Randise-Hinchliff C, Kinnings S, Deshmukh S, Alla N, Tan C, Kia A, Greene G, Leety L, Rhoa M, Yeats S, Saul M, Chou J, Bianco K, O'Shea K, Bujold E, Norwitz E, Wapner R, Saade G, Kaper F. Circulating transcripts in maternal blood reflect a molecular signature of early-onset preeclampsia. Sci Transl Med 2021; 12:12/550/eaaz0131. [PMID: 32611681 DOI: 10.1126/scitranslmed.aaz0131] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/07/2019] [Accepted: 06/08/2020] [Indexed: 12/13/2022]
Abstract
Circulating RNA (C-RNA) is continually released into the bloodstream from tissues throughout the body, offering an opportunity to noninvasively monitor all aspects of pregnancy health from conception to birth. We asked whether C-RNA analysis could robustly detect aberrations in patients diagnosed with preeclampsia (PE), a prevalent and potentially fatal pregnancy complication. As an initial examination, we sequenced the circulating transcriptome from 40 pregnancies at the time of severe, early-onset PE diagnosis and 73 gestational age-matched controls. Differential expression analysis identified 30 transcripts with gene ontology annotations and tissue expression patterns consistent with the placental dysfunction, impaired fetal development, and maternal immune and cardiovascular system dysregulation characteristic of PE. Furthermore, machine learning identified combinations of 49 C-RNA transcripts that classified an independent cohort of patients (early-onset PE, n = 12; control, n = 12) with 85 to 89% accuracy. C-RNA may thus hold promise for improving the diagnosis and identification of at-risk pregnancies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Emmanuel Bujold
- Department of Obstetrics and Gynecology and Department of Social and Preventive Medicine, Faculty of Medicine, Université Laval, Quebec City, Quebec G1V 086, Canada
| | - Errol Norwitz
- Department of Obstetrics and Gynecology and the Mother Infant Research Institute, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Ronald Wapner
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY 10032, USA
| | - George Saade
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | |
Collapse
|
21
|
Gannoun MBA, Raguema N, Zitouni H, Mehdi M, Seda O, Mahjoub T, Lavoie JL. MMP-2 and MMP-9 Polymorphisms and Preeclampsia Risk in Tunisian Arabs: A Case-Control Study. J Clin Med 2021; 10:jcm10122647. [PMID: 34208487 PMCID: PMC8234886 DOI: 10.3390/jcm10122647] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/03/2021] [Accepted: 06/10/2021] [Indexed: 01/18/2023] Open
Abstract
The abnormal production of matrix metalloproteinases (MMPs), especially MMP-9 and MMP-2, plays a pivotal role in hypertensive disorders of pregnancy, and as such, can influence the development of preeclampsia. These alterations may result from functional genetic polymorphisms in the promoter region of MMP-9 and MMP-2 genes, which modify MMP-9 and MMP-2 expression. We investigated the association of MMP-9 polymorphism rs3918242 (-1562 C>T) and MMP-2 polymorphism rs2285053 (-735 C>T) with the risk of preeclampsia. This case–control study was conducted on 345 women with preeclampsia and 281 age-matched women with normal pregnancies from Tunisian hospitals. Genomic DNA was extracted from whole blood collected at delivery. Genotypes for -1562 C>T and -735 C>T polymorphisms were performed using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). An increased frequency of heterozygous MMP-9 -1562 C/T genotype carriers was observed in women with preeclampsia compared to healthy controls (p = 0.03). In contrast, the MMP-2 -735 C>T polymorphism was not significantly different regarding frequency distribution of the allele and genotype between healthy pregnant women and women with preeclampsia. Our study suggests that the MMP-9 -1562 C/T variant, associated with high MMP-9 production, could be a genetic risk factor for preeclampsia in Tunisian women.
Collapse
Affiliation(s)
- Marwa Ben Ali Gannoun
- Laboratory of Human Genome and Multifactorial Diseases, Faculty of Pharmacy of Monastir, University of Monastir, Monastir 5000, Tunisia; (M.B.A.G.); (N.R.); (H.Z.); (T.M.)
- Laboratory of Histology Embryology and Cytogenetics, Faculty of Medicine, University of Monastir, Monastir 5000, Tunisia
| | - Nozha Raguema
- Laboratory of Human Genome and Multifactorial Diseases, Faculty of Pharmacy of Monastir, University of Monastir, Monastir 5000, Tunisia; (M.B.A.G.); (N.R.); (H.Z.); (T.M.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
- School of Kinesiology and Physical Activity Sciences, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Hedia Zitouni
- Laboratory of Human Genome and Multifactorial Diseases, Faculty of Pharmacy of Monastir, University of Monastir, Monastir 5000, Tunisia; (M.B.A.G.); (N.R.); (H.Z.); (T.M.)
| | - Meriem Mehdi
- Laboratory of Cytogenetics and Reproductive Biology, Center of Maternity and Neonatology Monastir, Fattouma Bourguiba University Teaching Hospital, Monastir 5000, Tunisia;
| | - Ondrej Seda
- The First Faculty of Medicine and General University Hospital, Institute of Biology and Medical Genetics, Charles University, 12800 Prague, Czech Republic;
| | - Touhami Mahjoub
- Laboratory of Human Genome and Multifactorial Diseases, Faculty of Pharmacy of Monastir, University of Monastir, Monastir 5000, Tunisia; (M.B.A.G.); (N.R.); (H.Z.); (T.M.)
| | - Julie L. Lavoie
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
- School of Kinesiology and Physical Activity Sciences, Université de Montréal, Montréal, QC H3T 1J4, Canada
- Correspondence: ; Tel.: +1-(514)-890-8000 (ext. 23612)
| |
Collapse
|
22
|
Subi TM, Krishnakumar V, Kataru CR, Panigrahi I, Kannan M. Association of VEGF and p53 Polymorphisms and Spiral Artery Remodeling in Recurrent Pregnancy Loss: A Systematic Review and Meta-Analysis. Thromb Haemost 2021; 122:363-376. [PMID: 34041737 DOI: 10.1055/a-1518-1756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Many studies have reported the association of VEGF-1154G/A, VEGF 936C/T, and p53 Arg72Pro polymorphisms with recurrent pregnancy loss (RPL), but the outcomes are inconsistent. We have used a meta-analysis to associate these polymorphisms with RPL, having the spiral artery remodeling as a major risk factor. The studies were identified from three different reputed databases, namely ScienceDirect, PubMed/Medline, and Scopus. The eligible studies of VEGF-1154G/A, VEGF 936C/T, and p53Arg72Pro polymorphisms associated with the RPL were selected for the analysis. They were segregated into three different ethnic groups as Asians, Caucasians, and mixed population. For the analysis, the overall prevalence, odds ratio, risk ratio, relative risk ratio, and p-values were calculated. A total of 3,241 RPL cases and 3,205 healthy controls from 21 different case-control studies were analyzed. RPL was highly prevalent in the mixed population with VEGF-1154G/A and p53 Arg72Pro polymorphisms (70.04 and 66.46%, respectively) and in the Asian population with VEGF 936C/T polymorphism (53.58%). The homozygous recessive genotypes of VEGF and p53 exhibited significant association between the respective polymorphisms and RPL along with the increased risk of outcome. The current analysis conclusively reports the geographic distribution of the different genetic polymorphisms which shows high association with the progression of RPL. Understanding the spectrum of polymorphisms on different populations with the spiral artery remodeling as a risk factor encloses the importance of the vasculature during the pregnancy.
Collapse
Affiliation(s)
- Tamil Mani Subi
- Division of Blood and Vascular Biology, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India
| | - Vinodhini Krishnakumar
- Division of Blood and Vascular Biology, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India
| | - Chandreswara Raju Kataru
- Division of Blood and Vascular Biology, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India
| | - Inusha Panigrahi
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Meganathan Kannan
- Division of Blood and Vascular Biology, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India
| |
Collapse
|
23
|
Galkin AP, Sysoev EI. Stress Response Is the Main Trigger of Sporadic Amyloidoses. Int J Mol Sci 2021; 22:4092. [PMID: 33920986 PMCID: PMC8071232 DOI: 10.3390/ijms22084092] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 12/11/2022] Open
Abstract
Amyloidoses are a group of diseases associated with the formation of pathological protein fibrils with cross-β structures. Approximately 5-10% of the cases of these diseases are determined by amyloidogenic mutations, as well as by transmission of infectious amyloids (prions) between organisms. The most common group of so-called sporadic amyloidoses is associated with abnormal aggregation of wild-type proteins. Some sporadic amyloidoses are known to be induced only against the background of certain pathologies, but in some cases the cause of amyloidosis is unclear. It is assumed that these diseases often occur by accident. Here we present facts and hypotheses about the association of sporadic amyloidoses with vascular pathologies, trauma, oxidative stress, cancer, metabolic diseases, chronic infections and COVID-19. Generalization of current data shows that all sporadic amyloidoses can be regarded as a secondary event occurring against the background of diseases provoking a cellular stress response. Various factors causing the stress response provoke protein overproduction, a local increase in the concentration or modifications, which contributes to amyloidogenesis. Progress in the treatment of vascular, metabolic and infectious diseases, as well as cancers, should lead to a significant reduction in the risk of sporadic amyloidoses.
Collapse
Affiliation(s)
- Alexey P. Galkin
- St. Petersburg Branch, Vavilov Institute of General Genetics, 199034 St. Petersburg, Russia
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia;
| | - Evgeniy I. Sysoev
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia;
| |
Collapse
|
24
|
Manley CN, Deepak V, Ravikumar N, Smith AK, Knight AK, Badell ML, Sidell N, Rajakumar A. Transcription factor AP2A affects sFLT1 expression and decidualization in decidual stromal cells: Implications to preeclampsia pathology. Pregnancy Hypertens 2020; 21:152-158. [PMID: 32535226 DOI: 10.1016/j.preghy.2020.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/01/2020] [Accepted: 06/05/2020] [Indexed: 12/21/2022]
Abstract
Preeclampsia (PE) yields a spectrum of phenotypic expression, leading to varying degrees of hypertension, maternal renal dysfunction and placental insufficiency with resultant maternal and neonatal morbidity. Increased sFLT1 expression contributing to angiogenic factor imbalance, placental hypoxia, failed immune adaptation to the fetus and defective decidualization are among the commonly proposed theories of PE pathogenesis. Recently researchers have focused their attention on the events that occur at the maternal fetal interface as potential contributors to PE pathogenesis. Decidual stromal cells (DSC) isolated from preeclamptic women show diminished ability to decidualize upon stimulation and reduced capacity to downregulate sFlt-1 levels. In this study, we sought to gain insight into the molecular mechanism(s) involved in the aberrant decidualization capacity of PE DSC. Our findings using qRT-PCR show that PE DSCs have 6-fold higher basal levels of transcription factor AP2A (TFAP2A) RNA compared to women without PE and that expression of TFAP2A increases during decidualization but only in DSCs of normotensive (NT) women. Silencing of TFAP2A using Trilencer siRNA upregulated sFLT1 expression only in NT-DSCs but suppressed the expression of decidualization markers PRL, IGFBP1 and their regulator FOXO1 in cells from both groups. Collectively, our observations suggest that TFAP2A acts as a repressor of sFLT1 and plays a necessary role in decidualization possibly through interacting with another factor that is aberrantly expressed in PE DSCs.
Collapse
Affiliation(s)
- Charisma N Manley
- Division of Maternal Fetal Medicine, Emory University School of Medicine, Atlanta, GA, United States; Department of Gynecology & Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Venkataraman Deepak
- Division of Research, Emory University School of Medicine, Atlanta, GA, United States; Department of Gynecology & Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Nithin Ravikumar
- Division of Research, Emory University School of Medicine, Atlanta, GA, United States; Department of Gynecology & Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Alicia K Smith
- Division of Research, Emory University School of Medicine, Atlanta, GA, United States; Department of Gynecology & Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Anna K Knight
- Division of Research, Emory University School of Medicine, Atlanta, GA, United States; Department of Gynecology & Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Martina L Badell
- Division of Maternal Fetal Medicine, Emory University School of Medicine, Atlanta, GA, United States; Department of Gynecology & Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Neil Sidell
- Division of Research, Emory University School of Medicine, Atlanta, GA, United States; Department of Gynecology & Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Augustine Rajakumar
- Division of Research, Emory University School of Medicine, Atlanta, GA, United States; Department of Gynecology & Obstetrics, Emory University School of Medicine, Atlanta, GA, United States.
| |
Collapse
|
25
|
Jadhav A, Khaire A, Joshi S. Exploring the role of oxidative stress, fatty acids and neurotrophins in gestational diabetes mellitus. Growth Factors 2020; 38:226-234. [PMID: 33703982 DOI: 10.1080/08977194.2021.1895143] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Gestational diabetes mellitus (GDM) constitutes an unfavorable intrauterine environment for embryonic and feto-placental development. Women with GDM are at higher risk for materno-fetal complications and placental abnormalities. The placenta acts as an interface between the maternal and fetal circulations and also plays an important role in protecting the fetus from adverse effects of maternal metabolic conditions. One of the earliest abnormalities observed in GDM pregnancies is increased oxidative stress in the placenta which affects fetal development. Imbalances in maternal nutrition particularly long-chain polyunsaturated fatty acid (LCPUFA) intake and/or metabolism lead to increased oxidative stress. Reports indicate that oxidative stress and LCPUFA such as docosahexaenoic acid affect the levels of neurotrophins. The present review aims to provide insights into a mechanistic link between oxidative stress, LCPUFA and neurotrophin in the placenta in women with GDM and its implications for neurodevelopmental outcomes in children.
Collapse
Affiliation(s)
- Anjali Jadhav
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Amrita Khaire
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Sadhana Joshi
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| |
Collapse
|
26
|
Chuva de Sousa Lopes SM, Alexdottir MS, Valdimarsdottir G. The TGFβ Family in Human Placental Development at the Fetal-Maternal Interface. Biomolecules 2020; 10:biom10030453. [PMID: 32183218 PMCID: PMC7175362 DOI: 10.3390/biom10030453] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023] Open
Abstract
Emerging data suggest that a trophoblast stem cell (TSC) population exists in the early human placenta. However, in vitro stem cell culture models are still in development and it remains under debate how well they reflect primary trophoblast (TB) cells. The absence of robust protocols to generate TSCs from humans has resulted in limited knowledge of the molecular mechanisms that regulate human placental development and TB lineage specification when compared to other human embryonic stem cells (hESCs). As placentation in mouse and human differ considerably, it is only with the development of human-based disease models using TSCs that we will be able to understand the various diseases caused by abnormal placentation in humans, such as preeclampsia. In this review, we summarize the knowledge on normal human placental development, the placental disease preeclampsia, and current stem cell model systems used to mimic TB differentiation. A special focus is given to the transforming growth factor-beta (TGFβ) family as it has been shown that the TGFβ family has an important role in human placental development and disease.
Collapse
Affiliation(s)
- Susana M. Chuva de Sousa Lopes
- Dept. Anatomy and Embryology, Leiden University Medical Center, 2300 Leiden, The Netherlands;
- Dept. Reproductive Medicine Anatomy and Embryology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Marta S. Alexdottir
- Department of Anatomy, BioMedical Center, University of Iceland, Sturlugata 8, 101 Reykjavik, Iceland;
| | - Gudrun Valdimarsdottir
- Department of Anatomy, BioMedical Center, University of Iceland, Sturlugata 8, 101 Reykjavik, Iceland;
- Correspondence: ; Tel.: +354-5254797
| |
Collapse
|
27
|
Pejkovska Ilieva M, Antovska V, Kaeva Pejkovska M. Uterine artery pulsatility index as a relevant parameter in the prediction of preeclampsia. MAKEDONSKO FARMACEVTSKI BILTEN 2020. [DOI: 10.33320/maced.pharm.bull.2020.66.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Preeclampsia is a clinical syndrome that occurs in 5-10% of pregnancies with increased perinatal morbidity and mortality. According to ISUOG (International Society of Ultrasound in Obstetrics & Gynecology), the use of the uterine artery pulsatility index (PI) is an important sensitive method in predicting the risk of preeclampsia.
This study is to emphasize the value of PI as a more relevant predictive parameter in the detection of preeclampsia in the second trimester compared to the presence of isolated uterine artery notch.
For the above purpose, 96 patients were examined at the University Clinic of Gynecology and Obstetrics in Skopje, divided into 2 groups: study and control group. The study group consisted of 48 patients from 14 to 20 gestational weeks with present uterine artery notch, being the main inclusion criterion. The control group consisted of 48 pregnant patients at the same gestational age with absence of uterine artery notch.
In the study group, 43.7% developed clinical syndrome of preeclampsia. The resistance index (RI) value was up to 0.73 and the predictive value of the RI was 57.1%. In patients who developed preeclampsia syndrome, the value of the PI above 1.75 was of much greater predictive value, with value of 71.4%. The sensitivity of these values was 88%, which is a reliable parameter.
PI is a reliable parameter in detecting the risk of developing preeclampsia and an indicator of great clinical significance in the daily practice in perinatology.
Keywords: preeclampsia; prediction; pulsatility index
Collapse
Affiliation(s)
- Maja Pejkovska Ilieva
- University Clinic for Obstetrics and Gynecology - Skopje, Ss. Cyril and Methodius University, Mother Theresa 17, 1000 Skopje, Republic of North Macedonia
| | - Vesna Antovska
- University Clinic for Obstetrics and Gynecology - Skopje, Ss. Cyril and Methodius University, Mother Theresa 17, 1000 Skopje, Republic of North Macedonia
| | - Mirjana Kaeva Pejkovska
- Institute of mental health for children and adolescent “Youth”- Skopje, Partizanski Odredi 60, 1000 Skopje, Republic of North Macedonia
| |
Collapse
|
28
|
Russell MW, Moldenhauer JS, Rychik J, Burnham NB, Zullo E, Parry SI, Simmons RA, Elovitz MA, Nicolson SC, Linn RL, Johnson MP, Yu S, Sampson MG, Hakonarson H, Gaynor JW. Damaging Variants in Proangiogenic Genes Impair Growth in Fetuses with Cardiac Defects. J Pediatr 2019; 213:103-109. [PMID: 31227283 PMCID: PMC6765419 DOI: 10.1016/j.jpeds.2019.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/30/2019] [Accepted: 05/09/2019] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To determine the impact of damaging genetic variation in proangiogenic pathways on placental function, complications of pregnancy, fetal growth, and clinical outcomes in pregnancies with fetal congenital heart defect. STUDY DESIGN Families delivering a baby with a congenital heart defect requiring surgical repair in infancy were recruited. The placenta and neonate were weighed and measured. Hemodynamic variables were recorded from a third trimester (36.4 ± 1.7 weeks) fetal echocardiogram. Exome sequencing was performed on the probands (N = 133) and consented parents (114 parent-child trios, and 15 parent-child duos) and the GeneVetter analysis tool used to identify damaging coding sequence variants in 163 genes associated with the positive regulation of angiogenesis (PRA) (GO:0045766). RESULTS In total, 117 damaging variants were identified in PRA genes in 133 congenital heart defect probands with 73 subjects having at least 1 variant. Presence of a damaging PRA variant was associated with increased umbilical artery pulsatility index (mean 1.11 with variant vs 1.00 without; P = .01). The presence of a damaging PRA variant was also associated with lower neonatal length and head circumference for age z score at birth (mean -0.44 and -0.47 with variant vs 0.23 and -0.05 without; P = .01 and .04, respectively). During median 3.1 years (IQR 2.0-4.1 years) of follow-up, deaths occurred in 2 of 60 (3.3%) subjects with no PRA variant and in 9 of 73 (12.3%) subjects with 1 or more PRA variants (P = .06). CONCLUSIONS Damaging variants in proangiogenic genes may impact placental function and are associated with impaired fetal growth in pregnancies involving a fetus with congenital heart defect.
Collapse
Affiliation(s)
- Mark W Russell
- Division of Pediatric Cardiology, Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI.
| | - Julie S Moldenhauer
- Center for Fetal Diagnosis and Therapy, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Jack Rychik
- Division of Pediatric Cardiology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Nancy B Burnham
- Division of Cardiothoracic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Erin Zullo
- Division of Cardiothoracic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Samuel I Parry
- Division of Maternal Fetal Medicine, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA
| | - Rebecca A Simmons
- Division of Neonatology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Michal A Elovitz
- Division of Maternal Fetal Medicine, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA
| | - Susan C Nicolson
- Division of Cardiothoracic Anesthesiology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Rebecca L Linn
- Division of Anatomic Pathology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Mark P Johnson
- Center for Fetal Diagnosis and Therapy, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Sunkyung Yu
- Division of Pediatric Cardiology, Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI
| | - Matthew G Sampson
- Division of Pediatric Nephrology, Department of Pediatrics, and Center for Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI
| | - Hakon Hakonarson
- The Center for Applied Genomics, The Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - J William Gaynor
- Division of Cardiothoracic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA
| |
Collapse
|
29
|
Kasture V, Sundrani D, Dalvi S, Swamy M, Kale A, Joshi S. Maternal omega-3 fatty acids and vitamin E improve placental angiogenesis in late-onset but not early-onset preeclampsia. Mol Cell Biochem 2019; 461:159-170. [PMID: 31420792 DOI: 10.1007/s11010-019-03599-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 08/08/2019] [Indexed: 02/08/2023]
Abstract
Abnormal placental vasculature is associated with preeclampsia. Preeclampsia is of two types, i.e., early- and late-onset preeclampsia (LOP), both having different etiologies. We have earlier demonstrated low levels of omega-3 fatty acids and vitamin E in women with preeclampsia. The current study examines the effect of maternal omega-3 fatty acids and vitamin E supplementation on angiogenic factors in a rat model of preeclampsia. Pregnant rats were divided into a total of five groups control, early-onset preeclampsia (EOP); LOP; EOP supplemented with omega-3 fatty acid and vitamin E and LOP supplemented with omega-3 fatty acid and vitamin E. Preeclampsia was induced by administering L-nitroarginine methylester (L-NAME) at the dose of 50 mg/kg body weight/day. The vascular endothelial growth factor gene expression and protein levels were lower (p < 0.01 for both) in animals from both EOP as well as LOP groups (p < 0.01). In the EOP group, the protein levels of VEGF receptor-1 were also lower (p < 0.01). Supplementation of omega-3 fatty acids and vitamin E to LOP improved the levels of VEGF and VEGF receptor-1 only in the LOP but not in the EOP group. In the EOP group, the gene expression of hypoxia inducible factor 1 alpha (HIF-1α) in the placenta was higher (p < 0.05) and supplementation normalized these levels. Our findings indicate that maternal supplementation of omega-3 fatty acids and vitamin E has differential effect on preeclampsia subtypes.
Collapse
Affiliation(s)
- Vaishali Kasture
- Department of Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, Pune-Satara Road, Pune, 411043, India
| | - Deepali Sundrani
- Department of Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, Pune-Satara Road, Pune, 411043, India
| | - Surabhi Dalvi
- Department of Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, Pune-Satara Road, Pune, 411043, India
| | - Mayur Swamy
- Department of Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, Pune-Satara Road, Pune, 411043, India
| | - Anvita Kale
- Department of Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, Pune-Satara Road, Pune, 411043, India
| | - Sadhana Joshi
- Department of Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, Pune-Satara Road, Pune, 411043, India.
| |
Collapse
|
30
|
Mochan S, Dhingra MK, Gupta SK, Saxena S, Arora P, Yadav V, Rani N, Luthra K, Dwivedi S, Bhatla N, Dhingra R. Status of VEGF in preeclampsia and its effect on endoplasmic reticulum stress in placental trophoblast cells. Eur J Obstet Gynecol Reprod Biol X 2019; 4:100070. [PMID: 31517301 PMCID: PMC6728727 DOI: 10.1016/j.eurox.2019.100070] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 05/18/2019] [Accepted: 06/11/2019] [Indexed: 11/19/2022] Open
Abstract
Objective To explore the role of VEGF in attenuating endoplasmic reticulum stress in placental trophoblast cells. Study design Study was divided into following parts: 1. Serum Analysis of GRP78 and VEGF using sandwich ELISA. 2. Expression of VEGF and GRP78 in placentae by immunohistochemistry (IHC). 3. In Vitro experiments. Status of ER stress markers (GRP78, eIF2α, XBP1, ATF6 and CHOP) was assessed at various time points (8 h, 14 h, 24 h) when trophoblast cells were treated with varying concentration(s) of VEGF and also by adding recombinant VEGF at protein (Immunofluorescence, Western blot) and transcript levels (qRT-PCR). Results Increased GRP78 and decreased VEGF protein levels in sera and placentae of preeclamptic pregnant women and reduced expression of various ER stress markers at both transcript and protein levels was observed in trophoblast cells when they were exposed to recombinant VEGF thereby indicating positive role of VEGF in alleviating ER stress. Conclusions Reduced expression of ER stress markers in trophoblast cells against increased VEGF highlighted a new window to explore prospective drugs that can be designed to modulate the activities of various ER stress sensors in order to alleviate ER stress in pregnant women with preeclampsia.
Collapse
Affiliation(s)
- Sankat Mochan
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
- Corresponding authors.
| | - Manoj Kumar Dhingra
- Department of Community Medicine, NC Medical college and hospital, Israna, Panipat, Haryana, 132107, India
| | - Sunil Kumar Gupta
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Shobhit Saxena
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Pallavi Arora
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Vineeta Yadav
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Neerja Rani
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Sadanand Dwivedi
- Department of Biostatistics, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Neerja Bhatla
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Renu Dhingra
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
- Corresponding authors.
| |
Collapse
|
31
|
Gaynor JW, Parry S, Moldenhauer JS, Simmons RA, Rychik J, Ittenbach RF, Russell WW, Zullo E, Ward JL, Nicolson SC, Spray TL, Johnson MP. The impact of the maternal-foetal environment on outcomes of surgery for congenital heart disease in neonates. Eur J Cardiothorac Surg 2019; 54:348-353. [PMID: 29447332 DOI: 10.1093/ejcts/ezy015] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 01/04/2018] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVES Pregnancies with congenital heart disease in the foetus have an increased prevalence of pre-eclampsia, small for gestational age and preterm birth, which are evidence of an impaired maternal-foetal environment (MFE). METHODS The impact of an impaired MFE, defined as pre-eclampsia, small for gestational age or preterm birth, on outcomes after cardiac surgery was evaluated in neonates (n = 135) enrolled in a study evaluating exposure to environmental toxicants and neuro-developmental outcomes. RESULTS The most common diagnoses were transposition of the great arteries (n = 47) and hypoplastic left heart syndrome (n = 43). Impaired MFE was present in 28 of 135 (21%) subjects, with small for gestational age present in 17 (61%) patients. The presence of an impaired MFE was similar for all diagnoses, except transposition of the great arteries (P < 0.006). Postoperative length of stay was shorter for subjects without an impaired MFE (14 vs 38 days, P < 0.001). Hospital mortality was not significantly different with or without impaired MFE (11.7% vs 2.8%, P = 0.104). However, for the entire cohort, survival at 36 months was greater for those without an impaired MFE (96% vs 68%, P = 0.001). For patients with hypoplastic left heart syndrome, survival was also greater for those without an impaired MFE (90% vs 43%, P = 0.007). CONCLUSIONS An impaired MFE is common in pregnancies in which the foetus has congenital heart disease. After cardiac surgery in neonates, the presence of an impaired MFE was associated with lower survival at 36 months of age for the entire cohort and for the subgroup with hypoplastic left heart syndrome.
Collapse
Affiliation(s)
- James William Gaynor
- Division of Cardiac Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Samuel Parry
- Division of Maternal Fetal Medicine, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA
| | - Julie S Moldenhauer
- Center for Fetal Diagnosis and Therapy, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rebecca A Simmons
- Division of Neonatology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jack Rychik
- Division of Pediatric Cardiology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Richard F Ittenbach
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - William W Russell
- Division of Cardiac Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Erin Zullo
- Division of Cardiac Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - John Laurenson Ward
- Division of Cardiac Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Susan C Nicolson
- Division of Pediatric Cardiac Anesthesiology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Thomas L Spray
- Division of Cardiac Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mark P Johnson
- Center for Fetal Diagnosis and Therapy, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
32
|
Zhang Y, Li P, Guo Y, Liu X, Zhang Y. MMP-9 and TIMP-1 in placenta of hypertensive disorder complicating pregnancy. Exp Ther Med 2019; 18:637-641. [PMID: 31258700 PMCID: PMC6566117 DOI: 10.3892/etm.2019.7591] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 04/19/2019] [Indexed: 12/26/2022] Open
Abstract
Expression and characteristics of matrix metalloproteinase-9 (MMP-9) and TIMP metallopeptidase inhibitor-1 (TIMP-1) in the placenta of pregnancy induced hypertension (PIH) were detected in Uygur to analyze its correlation with PIH, and to provide a theoretical basis for clinical work. Ninety cases of placental tissue patients who were hospitalized in the Department of Obstetrics of People's Hospital of Xinjiang Uygur from December 2014 to September 2016 were collected, including 30 cases of severe preeclampsia, 30 cases of mild preeclampsia, and 30 cases of normal group. The distribution of MMP-9 and TIMP-1 in placenta was mainly in the cytoplasm of trophoblast cells, vascular endothelial cells and villous mesenchymal cells. The distribution of MMP-9 positive particles in the placenta tissue of the severe group was significantly reduced. The difference of MMP-9 in the three groups was statistically significant. Severe group was statistically significantly different from normal and mild group. With the aggravation of PIH, positive expression of MMP-9 was gradually decreased. TIMP-1 was expressed in each group, and difference was not statistically significant. Positive expression ratio of MMP-9/TIMP-1 in severe group was lower than that in normal pregnancy and mild group, and positive expression ratio of the two groups became smaller as the condition worsened. Positive expression of MMP-9 in placental tissue of patients with PIH decreased significantly with the severity of PIH. TIMP-1 in placental tissue of PIH patients did not change much in each group, and had no significant correlation with PIH.
Collapse
Affiliation(s)
- Yuyi Zhang
- Department of Gynecology and Obstetrics, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei 441000, P.R. China
| | - Peng Li
- Department of General Surgery, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei 441000, P.R. China
| | - Yue Guo
- Department of Gynecology and Obstetrics, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei 441000, P.R. China
| | - Xiaowan Liu
- Department of Gynecology, People's Hospital of Xinjiang Uygur, Urumqi, Xinjiang Uygur Autonomous Region 830001, P.R. China
| | - Yubo Zhang
- Department of Stomatology, Quzhou No. 2 People's Hospital, Jinhua, Zhejiang 321000, P.R. China
| |
Collapse
|
33
|
Badran M, Abuyassin B, Ayas N, Laher I. Intermittent hypoxia impairs uterine artery function in pregnant mice. J Physiol 2019; 597:2639-2650. [PMID: 31002746 PMCID: PMC6826231 DOI: 10.1113/jp277775] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/27/2019] [Indexed: 01/05/2023] Open
Abstract
KEY POINTS Obstructive sleep apnoea (OSA) is a chronic condition characterized by intermittent hypoxia that induces oxidative stress and inflammation leading to cardiovascular disease. Women can develop OSA during late pregnancy, which is associated with adverse maternal and fetal outcomes. However, the effects of OSA throughout pregnancy on fetoplacental outcomes are unknown. Using a mouse model of intermittent hypoxia, we evaluated main uterine artery function, spiral artery remodelling, circulating angiogenic and anti-angiogenic factors, and placental hypoxia and oxidative stress at gestational day 14.5 in pregnant mice. Gestational intermittent hypoxia increased placental weight but decreased fetal weight, impaired uterine artery function, increased circulating angiogenic and anti-angiogenic factors, and induced placental hypoxia and oxidative stress, but had no impact on spiral artery remodelling. Our results suggest that pregnant women experiencing OSA during pregnancy could be at risk of maternal and fetal complications. ABSTRACT Obstructive sleep apnoea (OSA) is characterized by chronic intermittent hypoxia (IH) and is associated with increased inflammation, oxidative stress and endothelial dysfunction. OSA is a common sleep disorder and remains under-diagnosed; it can increase the risk of adverse maternal and fetal outcomes in pregnant women. We investigated the effects of gestational IH (GIH) on uterine artery function, spiral artery remodelling and placental circulating angiogenic and anti-angiogenic factors in pregnant female mice. WT C57BL/6 mice (8 weeks) were exposed to either GIH ( F I O 2 12%) or intermittent air ( F I O 2 21%) for 14.5 days of gestation. Exposure to GIH reduced fetal weight but increased placental weight. GIH dams had higher plasma levels of oxidative stress (8-isoprostane) and inflammatory markers (tumour necrosis factor-α). GIH significantly reduced uterine artery function as indicated by reduced endothelium-dependent vasodilatation and enhanced vasoconstriction. Plasma levels of placental angiogenic and anti-angiogenic markers (soluble fms-like tyrosine kinase-1, soluble endoglin, angiogenic placental growth factor-2 and vascular endothelial growth factor) were higher in pregnant mice exposed to GIH. There was no evidence of impaired spiral artery remodelling based on immunostaining with α-smooth muscle actin and cytokeratin-7, and also by measurements of lumen area. Immunostaining for markers of hypoxia (pimonidazole) and oxidative stress (4-hydroxynonenal) were higher in mice exposed to GIH. Our data show that GIH adversely affects uterine vascular function and may be a mechanism by which gestational OSA leads to adverse maternal and fetal outcomes.
Collapse
Affiliation(s)
- Mohammad Badran
- Department of AnesthesiologyPharmacology and TherapeuticsUniversity of British ColumbiaVancouverCanada
| | - Bisher Abuyassin
- Department of AnesthesiologyPharmacology and TherapeuticsUniversity of British ColumbiaVancouverCanada
| | - Najib Ayas
- Divisions of Critical Care and Respiratory MedicineDepartment of MedicineUniversity of British ColumbiaVancouverBCCanada
- Sleep Disorders ProgramUBC HospitalVancouverBCCanada
- Division of Critical Care MedicineProvidence HealthcareVancouverBCCanada
| | - Ismail Laher
- Department of AnesthesiologyPharmacology and TherapeuticsUniversity of British ColumbiaVancouverCanada
| |
Collapse
|
34
|
Tayyar AT, Karakus R, Eraslan Sahin M, Topbas NF, Sahin E, Karakus S, Yalcın ET, Tayyar A. Wnt signaling pathway in early- and late-onset preeclampsia: evaluation with Dickkopf-1 and R-Spondin-3 glycoproteins. Arch Gynecol Obstet 2019; 299:1551-1556. [DOI: 10.1007/s00404-019-05126-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/16/2019] [Indexed: 10/27/2022]
|
35
|
Mazibuko M, Moodley J, Naicker T. Dysregulation of circulating sTie2 and sHER2 in HIV-infected women with preeclampsia. Hypertens Pregnancy 2019; 38:89-95. [PMID: 30836796 DOI: 10.1080/10641955.2019.1584211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE This study assesses whether circulating sTie2 and sHER2 are altered in HIV-negative and HIV-positive pregnant normotensive and preeclamptic women. METHODS Serum samples were obtained from 80 pregnant women, stratified into four groups, namely, HIV-negative normotensives (20); HIV-positive normotensives (20); HIV-negative preeclamptics (20); and HIV-positive preeclamptics (20). The concentration of sTie2 and sHER2 was analyzed by Bio-Plex multiplex immunoassay and generated from a standard curve. RESULTS sTie2 differed significantly by pregnancy type (p = 0.0403) but not by HIV status (p = 0.5214). sHER2 did not show a significant difference between normotensive and preeclampsia (p = 0.3677) and by HIV status (p = 0.5249). CONCLUSION Irrespective of HIV status, reduced concentrations of sTie2 were evident in preeclampsia (PE) reflecting a dysregulation of the angiogenic process. sHER2 was similar between pregnancy types, attributable to the oxidative stressed microenvironment which promotes dysregulation of the MAPK and P13K/Akt signaling. HIV status did not influence sTie2 and sHER2 expression, reflecting the immune reconstitution of highly active antiretroviral therapy. sTie2 and sHER2 were not influenced by PE comorbid with HIV infection.
Collapse
Affiliation(s)
- Mduduzi Mazibuko
- a Optics and Imaging Centre, Nelson R. Mandela School of Medicine , University of KwaZulu-Natal , Durban , South Africa
| | - Jagidesa Moodley
- b Women's Health and HIV Research Group, Department of Obstetrics and Gynecology, Nelson R. Mandela School of Medicine , University of KwaZulu-Natal , Durban , KwaZulu-Natal , South Africa
| | - Thajasvarie Naicker
- a Optics and Imaging Centre, Nelson R. Mandela School of Medicine , University of KwaZulu-Natal , Durban , South Africa
| |
Collapse
|
36
|
Parchem JG, Kanasaki K, Kanasaki M, Sugimoto H, Xie L, Hamano Y, Lee SB, Gattone VH, Parry S, Strauss JF, Garovic VD, McElrath TF, Lu KH, Sibai BM, LeBleu VS, Carmeliet P, Kalluri R. Loss of placental growth factor ameliorates maternal hypertension and preeclampsia in mice. J Clin Invest 2018; 128:5008-5017. [PMID: 30179860 PMCID: PMC6205389 DOI: 10.1172/jci99026] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 08/28/2018] [Indexed: 12/28/2022] Open
Abstract
Preeclampsia remains a clinical challenge due to its poorly understood pathogenesis. A prevailing notion is that increased placental production of soluble fms-like tyrosine kinase-1 (sFlt-1) causes the maternal syndrome by inhibiting proangiogenic placental growth factor (PlGF) and VEGF. However, the significance of PlGF suppression in preeclampsia is uncertain. To test whether preeclampsia results from the imbalance of angiogenic factors reflected by an abnormal sFlt-1/PlGF ratio, we studied PlGF KO (Pgf-/-) mice and noted that the mice did not develop signs or sequelae of preeclampsia despite a marked elevation in circulating sFLT-1. Notably, PlGF KO mice had morphologically distinct placentas, showing an accumulation of junctional zone glycogen. We next considered the role of placental PlGF in an established model of preeclampsia (pregnant catechol-O-methyltransferase-deficient [COMT-deficient] mice) by generating mice with deletions in both the Pgf and Comt genes. Deletion of placental PlGF in the context of COMT loss resulted in a reduction in maternal blood pressure and increased placental glycogen, indicating that loss of PlGF might be protective against the development of preeclampsia. These results identify a role for PlGF in placental development and support a complex model for the pathogenesis of preeclampsia beyond an angiogenic factor imbalance.
Collapse
Affiliation(s)
- Jacqueline G Parchem
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas, USA
| | - Keizo Kanasaki
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Megumi Kanasaki
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Hikaru Sugimoto
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Liang Xie
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Yuki Hamano
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Soo Bong Lee
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Vincent H Gattone
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Samuel Parry
- Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jerome F Strauss
- Department of Obstetrics and Gynecology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Vesna D Garovic
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Thomas F McElrath
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Karen H Lu
- Department of Gynecologic Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Baha M Sibai
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Valerie S LeBleu
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Center for Cancer Biology (CCB), Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
37
|
Zou QY, Zhao YJ, Zhou C, Liu AX, Zhong XQ, Yan Q, Li Y, Yi FX, Bird IM, Zheng J. G Protein α Subunit 14 Mediates Fibroblast Growth Factor 2-Induced Cellular Responses in Human Endothelial Cells. J Cell Physiol 2018; 234:10184-10195. [PMID: 30387149 DOI: 10.1002/jcp.27688] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/09/2018] [Indexed: 12/20/2022]
Abstract
During pregnancy, a tremendous increase in fetoplacental angiogenesis is associated with elevated blood flow. Aberrant fetoplacental vascular function may lead to pregnancy complications including pre-eclampsia. Fibroblast growth factor 2 (FGF2) and vascular endothelial growth factor A (VEGFA) are crucial regulators of fetoplacental endothelial function. G protein α subunit 14 (GNA14), a member of Gαq/11 subfamily is involved in mediating hypertensive diseases and tumor vascularization. However, little is known about roles of GNA14 in mediating the FGF2- and VEGFA-induced fetoplacental endothelial function. Using human umbilical vein endothelial cells (HUVECs) cultured under physiological chronic low oxygen (3% O2 ) as a cell model, we show that transfecting cells with adenovirus carrying GNA14 complementary DNA (cDNA; Ad-GNA14) increases (p < 0.05) protein expression of GNA14. GNA14 overexpression blocks (p < 0.05) FGF2-stimulated endothelial migration, whereas it enhances (p < 0.05) endothelial monolayer integrity (maximum increase of ~35% over the control at 24 hr) in response to FGF2. In contrast, GNA14 overexpression does not significantly alter VEGFA-stimulated cell migration, VEGFA-weakened cell monolayer integrity, and intracellular Ca++ mobilization in response to adenosine triphosphate (ATP), FGF2, and VEGFA. GNA14 overexpression does not alter either FGF2- or VEGFA-induced phosphorylation of ERK1/2. However, GNA14 overexpression time-dependently elevates (p < 0.05) phosphorylation of phospholipase C-β3 (PLCβ3) at S1105 in response to FGF2, but not VEGFA. These data suggest that GNA14 distinctively mediates fetoplacental endothelial cell migration and permeability in response to FGF2 and VEGFA, possibly in part by altering activation of PLCβ3 under physiological chronic low oxygen.
Collapse
Affiliation(s)
- Qing-Yun Zou
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Ying-Jie Zhao
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Rheumatology, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Chi Zhou
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Ai-Xia Liu
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Reproductive Endocrinology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xin-Qi Zhong
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qin Yan
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yan Li
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Fu-Xian Yi
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Ian M Bird
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin.,Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
38
|
West RC, Bouma GJ, Winger QA. Shifting perspectives from "oncogenic" to oncofetal proteins; how these factors drive placental development. Reprod Biol Endocrinol 2018; 16:101. [PMID: 30340501 PMCID: PMC6195737 DOI: 10.1186/s12958-018-0421-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/09/2018] [Indexed: 12/23/2022] Open
Abstract
Early human placental development strongly resembles carcinogenesis in otherwise healthy tissues. The progenitor cells of the placenta, the cytotrophoblast, rapidly proliferate to produce a sufficient number of cells to form an organ that will contribute to fetal development as early as the first trimester. The cytotrophoblast cells begin to differentiate, some towards the fused cells of the syncytiotrophoblast and some towards the highly invasive and migratory extravillous trophoblast. Invasion and migration of extravillous trophoblast cells mimics tumor metastasis. One key difference between cancer progression and placental development is the tight regulation of these oncogenes and oncogenic processes. Often, tumor suppressors and oncogenes work synergistically to regulate cell proliferation, differentiation, and invasion in a restrained manner compared to the uncontrollable growth in cancer. This review will compare and contrast the mechanisms that drive both cancer progression and placental development. Specifically, this review will focus on the molecular mechanisms that promote cell proliferation, evasion of apoptosis, cell invasion, and angiogenesis.
Collapse
Affiliation(s)
- Rachel C. West
- Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory, Colorado State University, 10290 Ridgegate Circle, Lone Tree, Fort Collins, CO 80124 USA
| | - Gerrit J. Bouma
- Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory, Colorado State University, 10290 Ridgegate Circle, Lone Tree, Fort Collins, CO 80124 USA
| | - Quinton A. Winger
- Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory, Colorado State University, 10290 Ridgegate Circle, Lone Tree, Fort Collins, CO 80124 USA
| |
Collapse
|
39
|
Zitouni H, Ben Ali Gannoum M, Raguema N, Maleh W, Zouari I, Faleh RE, Guibourdenche J, Almawi WY, Mahjoub T. Contribution of angiotensinogen M235T and T174M gene variants and haplotypes to preeclampsia and its severity in (North African) Tunisians. J Renin Angiotensin Aldosterone Syst 2018; 19:1470320317753924. [PMID: 29366364 PMCID: PMC5843851 DOI: 10.1177/1470320317753924] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Preeclampsia (PE) is a pregnancy-associated hypertensive disorder and a leading cause of maternal and neonatal morbidity and mortality. While its pathogenesis remains ill defined, several candidate genes for PE have been identified, but results remain inconclusive. We investigated the association of the angiotensinogen ( AGT) gene variants M235T and T174M with PE, and we analyzed the contribution of both variants to the severity of PE. METHODS This case-control study enrolled 550 Tunisian pregnant women: 272 with PE, of whom 147 presented with mild, and 125 with severe PE, along with 278 unrelated age- and ethnically matched control women. AGT genotyping was performed by polymerase chain reaction-restriction fragment length polymorphism. RESULTS Significantly higher M235T minor allele frequency (MAF) was associated with increased risk of PE ( p < 0.001). Decreased frequency of heterozygous T174M genotype carriers were found in control women ( p = 0.015), suggesting a protective effect of this genotype (odds ratio (95% confidence interval) = 0.51 (0.29-0.89)). Two-locus haplotype analysis demonstrated MM and TT haplotypes to be negatively and positively associated with PE, respectively. MAF of M253T, but not T174M, was higher in the severe PE group, and carrying M235T or T174M minor allele was associated with increased body mass index ( p < 0.001) among unselected PE women. CONCLUSIONS AGT M235T and T174M variants contribute to an increased risk of developing PE, and for M235T to PE severity.
Collapse
Affiliation(s)
- Hedia Zitouni
- 1 Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), College of Pharmacy, University of Monastir, Tunisia.,2 Faculty of Science of Bizerte, University of Carthage, Tunisia.,3 INSERM UMR-S1139 College of Pharmacy, Paris Descartes University, France
| | - Marwa Ben Ali Gannoum
- 1 Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), College of Pharmacy, University of Monastir, Tunisia.,2 Faculty of Science of Bizerte, University of Carthage, Tunisia
| | - Nozha Raguema
- 1 Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), College of Pharmacy, University of Monastir, Tunisia.,2 Faculty of Science of Bizerte, University of Carthage, Tunisia
| | - Wided Maleh
- 4 Centre of Maternity and Neonatology, Tunisia
| | - Ines Zouari
- 4 Centre of Maternity and Neonatology, Tunisia
| | | | - Jean Guibourdenche
- 3 INSERM UMR-S1139 College of Pharmacy, Paris Descartes University, France
| | - Wassim Y Almawi
- 5 Faculty of Science of Tunis, University of Tunis El Manar, Tunisia
| | - Touhami Mahjoub
- 1 Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), College of Pharmacy, University of Monastir, Tunisia
| |
Collapse
|
40
|
Thakoordeen S, Moodley J, Naicker T. Candidate Gene, Genome-Wide Association and Bioinformatic Studies in Pre-eclampsia: a Review. Curr Hypertens Rep 2018; 20:91. [PMID: 30159611 DOI: 10.1007/s11906-018-0891-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Regardless of the familial linkage reported in pre-eclampsia development, understanding the polymorphic genes associated with pre-eclampsia remains limited. Hence, this review aims to outline the main genetic factors that have been investigated in respect to pre-eclampsia development. RECENT FINDINGS It is apparent that different genes show significance in varying populations. Notably, it is reported that apolipoprotein-1 gene polymorphisms are associated with pre-eclampsia development in an African-American population, which may be worthwhile to investigate in a Black South African cohort. Despite the research attention that is focused on this surreptitious syndrome, a definitive cause eludes scientists and physicians, alike. Genetic studies can fulfil a dual purpose of suggesting novel hypotheses through genome-wide screening and testing these hypotheses via candidate gene studies. However, publications to date have only presented inconsistent and conflicting results regarding candidate gene analysis.
Collapse
Affiliation(s)
- Semone Thakoordeen
- Women's Health and HIV Research Group, Department of Obstetrics and Gynaecology, University of KwaZulu-Natal, P Bag 7, Congella, KwaZulu-Natal, 4013, South Africa.
| | - Jagidesa Moodley
- Women's Health and HIV Research Group, Department of Obstetrics and Gynaecology, University of KwaZulu-Natal, P Bag 7, Congella, KwaZulu-Natal, 4013, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, University of KwaZulu-Natal, KwaZulu-Natal, South Africa
| |
Collapse
|
41
|
Geldenhuys J, Rossouw TM, Lombaard HA, Ehlers MM, Kock MM. Disruption in the Regulation of Immune Responses in the Placental Subtype of Preeclampsia. Front Immunol 2018; 9:1659. [PMID: 30079067 PMCID: PMC6062603 DOI: 10.3389/fimmu.2018.01659] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 07/04/2018] [Indexed: 01/21/2023] Open
Abstract
Preeclampsia is a pregnancy-specific disorder, of which one of its major subtypes, the placental subtype is considered a response to an ischemic placental environment, impacting fetal growth and pregnancy outcome. Inflammatory immune responses have been linked to metabolic and inflammatory disorders as well as reproductive failures. In healthy pregnancy, immune regulatory mechanisms prevent excessive systemic inflammation. However, in preeclampsia, the regulation of immune responses is disrupted as a result of aberrant activation of innate immune cells and imbalanced differentiation of T-helper cell subsets creating a cytotoxic environment in utero. Recognition events that facilitate immune interaction between maternal decidual T cells, NK cells, and cytotrophoblasts are considered an indirect cause of the incomplete remodeling of spiral arteries in preeclampsia. The mechanisms involved include the activation of immune cells and the subsequent secretion of cytokines and placental growth factors affecting trophoblast invasion, angiogenesis, and eventually placentation. In this review, we focus on the role of excessive systemic inflammation as the result of a dysregulated immune system in the development of preeclampsia. These include insufficient control of inflammation, failure of tolerance toward paternal antigens at the fetal-maternal interface, and subsequent over- or insufficient activation of immune mediators. It is also possible that external stimuli, such as bacterial endotoxin, may contribute to the excessive systemic inflammation in preeclampsia by stimulating the release of pro-inflammatory cytokines. In conclusion, a disrupted immune system might be a predisposing factor or result of placental oxidative stress or excessive inflammation in preeclampsia. Preeclampsia can thus be considered a hyperinflammatory state associated with defective regulation of the immune system proposed as a key element in the pathological events of the placental subtype of this disorder.
Collapse
Affiliation(s)
- Janri Geldenhuys
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Theresa Marie Rossouw
- Department of Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| | - Hendrik Andries Lombaard
- Obstetrics and Gynecology, Rahima Moosa Mother and Child Hospital, Wits Obstetrics and Gynecology Clinical Research Division, Faculty of Health Sciences, School of Clinical Medicine, University of Witwatersrand, Johannesburg, South Africa
| | - Marthie Magdaleen Ehlers
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Medical Microbiology, Tshwane Academic Division, National Health Laboratory Service, Pretoria, South Africa
| | - Marleen Magdalena Kock
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Medical Microbiology, Tshwane Academic Division, National Health Laboratory Service, Pretoria, South Africa
| |
Collapse
|
42
|
Monteiro LJ, Cubillos S, Sanchez M, Acuña-Gallardo S, Venegas P, Herrera V, Lam EWF, Varas-Godoy M, Illanes SE. Reduced FOXM1 Expression Limits Trophoblast Migration and Angiogenesis and Is Associated With Preeclampsia. Reprod Sci 2018; 26:580-590. [PMID: 29848205 DOI: 10.1177/1933719118778798] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Trophoblast cells are often compared to highly invasive carcinoma cells due to their capacity to proliferate in hypoxic conditions and to exhibit analogous vascular, proliferative, migratory, and invasive capacities. Thus, genes that are important for tumorigenesis, such as forkhead box M1 ( FOXM1) may also be involved in processes of trophoblast invasion. Indeed, we found Foxm1 protein and messenger RNA (mRNA) levels decreased as gestational age increased in rat's placentae. Accordingly, when mimicking early placental events in vitro, protein and mRNA expression of FOXM1 increased from 21% to 8% O2, reaching its highest expression at 3% oxygen tension, which reflects early implantation environment, and dropping to very low levels at 1% O2. Remarkably, FOXM1 silencing in JEG-3 cells was able to significantly decrease migration by 27.9%, in comparison with those cells transfected with control siRNA. Moreover, angiogenesis was compromised when conditioned media (CM) from FOXM1-siRNA -JEG-3 (3% O2) was added to human umbilical vein endothelial cells (HUVEC) cells; however, when CM of JEG-3 cells overexpressing FOXM1 at 1% O2 was added, the ability of HUVEC to form tubule networks was restored. Additionally, quantitative real-time polymerase chain reaction (PCR) assays of FOXM1 knockdown and overexpression experiments in JEG-3 cells revealed that the depletion of FOXM1 at 3% O2 and overexpression of FOXM1 at 1% O2 led to downregulation and upregulation of vascular endothelial growth factor transcriptional (VEGF) levels, respectively. Conversely, we also observed deregulation of FOXM1 in placentae derived from pregnancies complicated by preeclampsia (PE). Therefore, we demonstrate that FOXM1 may be a new regulatory protein of early placentation processes and that under chronic hypoxic conditions (1% O2) and in patients with severe PE, its levels decrease.
Collapse
Affiliation(s)
- Lara J Monteiro
- 1 Laboratory of Reproductive Biology, Centre for Biomedical Research, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Sofia Cubillos
- 1 Laboratory of Reproductive Biology, Centre for Biomedical Research, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Marianela Sanchez
- 1 Laboratory of Reproductive Biology, Centre for Biomedical Research, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Stephanie Acuña-Gallardo
- 1 Laboratory of Reproductive Biology, Centre for Biomedical Research, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Pía Venegas
- 1 Laboratory of Reproductive Biology, Centre for Biomedical Research, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,2 Department of Maternal-Foetal Medicine, Clínica Dávila, Santiago, Chile
| | - Valentina Herrera
- 1 Laboratory of Reproductive Biology, Centre for Biomedical Research, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Eric W-F Lam
- 3 Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Manuel Varas-Godoy
- 1 Laboratory of Reproductive Biology, Centre for Biomedical Research, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Sebastián E Illanes
- 1 Laboratory of Reproductive Biology, Centre for Biomedical Research, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,2 Department of Maternal-Foetal Medicine, Clínica Dávila, Santiago, Chile
| |
Collapse
|
43
|
Vaughan OR, Rossi CA, Ginsberg Y, White A, Hristova M, Sebire NJ, Martin J, Zachary IC, Peebles DM, David AL. Perinatal and long-term effects of maternal uterine artery adenoviral VEGF-A165 gene therapy in the growth-restricted guinea pig fetus. Am J Physiol Regul Integr Comp Physiol 2018; 315:R344-R353. [PMID: 29847165 DOI: 10.1152/ajpregu.00210.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Uterine artery application of adenoviral vascular endothelial growth factor A165 (Ad.VEGF-A165) gene therapy increases uterine blood flow and fetal growth in experimental animals with fetal growth restriction (FGR). Whether Ad.VEGF-A165 reduces lifelong cardiovascular disease risk imposed by FGR remains unknown. Here, pregnant guinea pigs fed 70% normal food intake to induce FGR received Ad.VEGF-A165 (1×1010 viral particles, n = 15) or vehicle ( n = 10), delivered to the external surface of the uterine arteries, in midpregnancy. Ad libitum-fed controls received vehicle only ( n = 14). Litter size, gestation length, and perinatal mortality were similar in control, untreated FGR, and FGR+Ad.VEGF-A165 animals. When compared with controls, birth weight was lower in male but higher in female pups following maternal nutrient restriction, whereas both male and female FGR+Ad.VEGF-A165 pups were heavier than untreated FGR pups ( P < 0.05, ANOVA). Postnatal weight gain was 10-20% greater in female FGR+Ad.VEGF-A165 than in untreated FGR pups, depending on age, although neither group differed from controls. Maternal nutrient restriction reduced heart weight in adult female offspring irrespective of Ad.VEGF-A165 treatment but did not alter ventricular wall thickness. In males, postnatal weight gain and heart morphology were not affected by maternal treatment. Neither systolic, diastolic, mean arterial pressure, adrenal weight, nor basal or challenged plasma cortisol were affected by maternal undernutrition or Ad.VEGF-A165 in either sex. Therefore, increased fetal growth conferred by maternal uterine artery Ad.VEGF-A165 is sustained postnatally in FGR female guinea pigs. In this study, we did not find evidence for an effect of maternal nutrient restriction or Ad.VEGF-A165 therapy on adult offspring blood pressure.
Collapse
Affiliation(s)
- O R Vaughan
- Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London , London , United Kingdom
| | - C A Rossi
- Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London , London , United Kingdom
| | - Y Ginsberg
- Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London , London , United Kingdom
| | - A White
- Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London , London , United Kingdom
| | - M Hristova
- Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London , London , United Kingdom
| | - N J Sebire
- Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London , London , United Kingdom
| | - J Martin
- Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London , London , United Kingdom
| | - I C Zachary
- Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London , London , United Kingdom
| | - D M Peebles
- Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London , London , United Kingdom
| | - A L David
- Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London , London , United Kingdom
| |
Collapse
|
44
|
Chiu YH, Yang MR, Wang LJ, Chen MH, Chang GD, Chen H. New insights into the regulation of placental growth factor gene expression by the transcription factors GCM1 and DLX3 in human placenta. J Biol Chem 2018; 293:9801-9811. [PMID: 29743241 DOI: 10.1074/jbc.ra117.001384] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 05/01/2018] [Indexed: 12/14/2022] Open
Abstract
Expression of placental growth factor (PGF) is closely associated with placental perfusion in early pregnancy. PGF is primarily expressed in placental trophoblasts, and its expression decreases in preeclampsia, associated with placental hypoxia. The transcription factors glial cells missing 1 (GCM1) and metal-regulatory transcription factor 1 (MTF1) have been implicated in the regulation of PGF gene expression through regulatory elements upstream and downstream of the PGF transcription start site, respectively. Here, we clarified the mechanism underlying placenta-specific PGF expression. We demonstrate that GCM1 up-regulates PGF expression through three downstream GCM1-binding sites (GBSs) but not a previously reported upstream GBS. Interestingly, we also found that these downstream GBSs also harbor metal-response elements for MTF1. Surprisingly, however, we observed that MTF1 is unlikely to regulate PGF expression in the placenta because knockdown or overexpression of GCM1, but not MTF1, dramatically decreased PGF expression or reversed the suppression of PGF expression under hypoxia, respectively. We also demonstrate that another transcription factor, Distal-less homeobox 3 (DLX3), interacts with the DNA-binding domain and the first transactivation domain of GCM1 and that this interaction inhibits GCM1-mediated PGF expression. Moreover, the GCM1-DLX3 interaction interfered with CREB-binding protein-mediated GCM1 acetylation and activation. In summary, we have identified several GBSs in the PGF promoter that are highly responsive to GCM1, have demonstrated that MTF1 does not significantly regulate PGF expression in placental cells, and provide evidence that DLX3 inhibits GCM1-mediated PGF expression. Our findings revise the mechanism for GCM1- and DLX3-mediated regulation of PGF gene expression.
Collapse
Affiliation(s)
- Yueh-Ho Chiu
- From the Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei 115, Taiwan and
| | - Ming-Ren Yang
- Graduate Institute of Biochemical Sciences, National Taiwan University, Taipei 106, Taiwan
| | - Liang-Jie Wang
- From the Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei 115, Taiwan and
| | - Ming-Hon Chen
- Graduate Institute of Biochemical Sciences, National Taiwan University, Taipei 106, Taiwan
| | - Geen-Dong Chang
- Graduate Institute of Biochemical Sciences, National Taiwan University, Taipei 106, Taiwan
| | - Hungwen Chen
- From the Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei 115, Taiwan and .,Graduate Institute of Biochemical Sciences, National Taiwan University, Taipei 106, Taiwan
| |
Collapse
|
45
|
Logue OC, Mahdi F, Chapman H, George EM, Bidwell GL. A Maternally Sequestered, Biopolymer-Stabilized Vascular Endothelial Growth Factor (VEGF) Chimera for Treatment of Preeclampsia. J Am Heart Assoc 2017; 6:e007216. [PMID: 29629873 PMCID: PMC5779036 DOI: 10.1161/jaha.117.007216] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/30/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Preeclampsia is a hypertensive syndrome that complicates 3% to 5% of pregnancies in the United States. Preeclampsia originates from an improperly vascularized and ischemic placenta that releases factors that drive systemic pathophysiology. One of these factors, soluble fms-like tyrosine kinase-1, is believed to sequester vascular endothelial growth factor (VEGF), leading to systemic endothelial dysfunction and hypertension. With the goal of targeting soluble fms-like tyrosine kinase-1 while simultaneously preventing fetal exposure to VEGF, we fused VEGF to elastin-like polypeptide, a biopolymer carrier that does not cross the placental barrier (ELP-VEGF). METHODS AND RESULTS ELP-VEGF restored in vitro endothelial cell tube formation in the presence of plasma from placental ischemic rats. Long-term administered ELP-VEGF in pregnant rats accumulated in maternal kidneys, aorta, liver, and placenta, but the protein was undetectable in the pups when administered at therapeutic doses in dams. Long-term administration of ELP-VEGF in a placental ischemia rat model achieved dose-dependent attenuation of hypertension, with blood pressure equal to sham controls at a dose of 5 mg/kg per day. ELP-VEGF infusion increased total plasma soluble fms-like tyrosine kinase-1 levels but dramatically reduced free plasma soluble fms-like tyrosine kinase-1 and induced urinary excretion of nitrate/nitrite, indicating enhanced renal nitric oxide signaling. ELP-VEGF at up to 5 mg/kg per day had no deleterious effect on maternal or fetal body weight. However, dose-dependent adverse events were observed, including ascites production and neovascular tissue encapsulation around the minipump. CONCLUSIONS ELP-VEGF has the potential to treat the preeclampsia maternal syndrome, but careful dosing and optimization of the delivery route are necessary.
Collapse
Affiliation(s)
- Omar C Logue
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS
| | - Fakhri Mahdi
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS
| | - Heather Chapman
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
| | - Eric M George
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS
| | - Gene L Bidwell
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
46
|
Helmo FR, Lopes AMM, Carneiro ACDM, Campos CG, Silva PB, Dos Reis Monteiro MLG, Rocha LP, Dos Reis MA, Etchebehere RM, Machado JR, Corrêa RRM. Angiogenic and antiangiogenic factors in preeclampsia. Pathol Res Pract 2017; 214:7-14. [PMID: 29174227 DOI: 10.1016/j.prp.2017.10.021] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Pre-eclampsia is a multifactorial hypertensive disorder that is triggered by placental insufficiency and that accounts for up to 15% of maternal deaths. In normal pregnancies, this process depends on the balance between the expression of angiogenic factors and antiangiogenic factors, which are responsible for remodeling the spiral arteries, as well as for neoangiogenesis and fetal development. PURPOSE The aim of this review is to discuss the main scientific findings regarding the role of angiogenic and antiangiogenic factors in the etiopathogenesis of preeclampsia. METHODS An extensive research was conducted in the Pubmed database in search of scientific manuscripts discussing potential associations between angiogenic and antiangiogenic factors and preeclampsia. Ninety-one papers were included in this review. RESULTS There is an increased expression of soluble fms-like tyrosine kinase receptor and soluble endoglin in pre-eclampsia, as well as reduced placental expression of vascular endothelial growth factor and placental growth factor. Systemic hypertension, proteinuria and kidney injury - such as enlargement and glomerular fibrin deposit, capillary occlusion due to edema, and hypertrophy of endocapillary cells - are some of these changes. The complex etiopathogenesis of preeclampsia instigates research of different biomarkers that allow for the early diagnosis of this entity, such as vascular endothelial growth factor, placental growth factor, soluble fms-like tyrosine kinase receptor, soluble endoglin, placental glycoprotein pregnancy-associated plasma protein-A and protein 13. CONCLUSION Even though it is possible to establish an efficient and effective diagnostic tool, three key principles must be observed in the management of preeclampsia: prevention, early screening and treatment.
Collapse
Affiliation(s)
- Fernanda Rodrigues Helmo
- Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Angela Maria Moed Lopes
- Oncology Research Institute, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Anna Cecília Dias Maciel Carneiro
- Discipline of Histology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro. Uberaba, Minas Gerais, Brazil
| | - Carolina Guissoni Campos
- Oncology Research Institute, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Polyana Barbosa Silva
- Oncology Research Institute, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | | | - Laura Penna Rocha
- Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Marlene Antônia Dos Reis
- Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Renata Margarida Etchebehere
- Surgical Pathology Service, Clinical Hospital, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Juliana Reis Machado
- Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil; Department of General Pathology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Rosana Rosa Miranda Corrêa
- Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil.
| |
Collapse
|
47
|
Li Y, Wang K, Zou QY, Jiang YZ, Zhou C, Zheng J. ITE Suppresses Angiogenic Responses in Human Artery and Vein Endothelial Cells: Differential Roles of AhR. Reprod Toxicol 2017; 74:181-188. [PMID: 28986273 DOI: 10.1016/j.reprotox.2017.09.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/22/2017] [Accepted: 09/27/2017] [Indexed: 12/26/2022]
Abstract
Aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor is involved in regulation of many essential biological processes including vascular development and angiogenesis. 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE) is an AhR ligand, which regulates immune responses and cancer cell growth. However, the roles of the ITE/AhR pathway in mediating placental angiogenesis remains elusive. Here, we determined if ITE affected placental angiogenic responses via AhR in human umbilical vein (HUVECs) and artery endothelial (HUAECs) cells in vitro. We observed that ITE dose- and time-dependently inhibited proliferation and viability of HUAECs and HUVECs, whereas it inhibited migration of HUAECs, but not HUVECs. While AhR siRNA significantly suppressed AhR protein expression in HUVECs and HUAECs, it attenuated the ITE-inhibited angiogenic responses of HUAECs, but not HUVECs. Collectively, ITE suppressed angiogenic responses of HUAECs and HUVECs, dependent and independent of AhR, respectively. These data suggest that ITE may regulate placental angiogenesis.
Collapse
Affiliation(s)
- Yan Li
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, United States
| | - Kai Wang
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, United States
| | - Qing-Yun Zou
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, United States
| | - Yi-Zhou Jiang
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, United States
| | - Chi Zhou
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, United States
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, United States; Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China.
| |
Collapse
|
48
|
McGinnis R, Steinthorsdottir V, Williams NO, Thorleifsson G, Shooter S, Hjartardottir S, Bumpstead S, Stefansdottir L, Hildyard L, Sigurdsson JK, Kemp JP, Silva GB, Thomsen LCV, Jääskeläinen T, Kajantie E, Chappell S, Kalsheker N, Moffett A, Hiby S, Lee WK, Padmanabhan S, Simpson NAB, Dolby VA, Staines-Urias E, Engel SM, Haugan A, Trogstad L, Svyatova G, Zakhidova N, Najmutdinova D, Dominiczak AF, Gjessing HK, Casas JP, Dudbridge F, Walker JJ, Pipkin FB, Thorsteinsdottir U, Geirsson RT, Lawlor DA, Iversen AC, Magnus P, Laivuori H, Stefansson K, Morgan L. Variants in the fetal genome near FLT1 are associated with risk of preeclampsia. Nat Genet 2017; 49:1255-1260. [PMID: 28628106 DOI: 10.1038/ng.3895] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 05/12/2017] [Indexed: 12/14/2022]
Abstract
Preeclampsia, which affects approximately 5% of pregnancies, is a leading cause of maternal and perinatal death. The causes of preeclampsia remain unclear, but there is evidence for inherited susceptibility. Genome-wide association studies (GWAS) have not identified maternal sequence variants of genome-wide significance that replicate in independent data sets. We report the first GWAS of offspring from preeclamptic pregnancies and discovery of the first genome-wide significant susceptibility locus (rs4769613; P = 5.4 × 10-11) in 4,380 cases and 310,238 controls. This locus is near the FLT1 gene encoding Fms-like tyrosine kinase 1, providing biological support, as a placental isoform of this protein (sFlt-1) is implicated in the pathology of preeclampsia. The association was strongest in offspring from pregnancies in which preeclampsia developed during late gestation and offspring birth weights exceeded the tenth centile. An additional nearby variant, rs12050029, associated with preeclampsia independently of rs4769613. The newly discovered locus may enhance understanding of the pathophysiology of preeclampsia and its subtypes.
Collapse
Affiliation(s)
| | | | | | | | | | - Sigrun Hjartardottir
- Department of Obstetrics and Gynecology, Landspitali University Hospital, Reykjavik, Iceland
| | | | | | | | | | - John P Kemp
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Gabriela B Silva
- Centre of Molecular Inflammation Research (CEMIR) and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Liv Cecilie V Thomsen
- Centre of Molecular Inflammation Research (CEMIR) and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Tiina Jääskeläinen
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Eero Kajantie
- National Institute for Health and Welfare, Helsinki, Finland
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- PEDEGO Research Unit, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Sally Chappell
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Noor Kalsheker
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Ashley Moffett
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Susan Hiby
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Wai Kwong Lee
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Sandosh Padmanabhan
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Nigel A B Simpson
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Leeds, UK
| | - Vivien A Dolby
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Leeds, UK
| | - Eleonora Staines-Urias
- Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
- Nuffield Department of Obstetrics &Gynaecology, University of Oxford, Oxford, UK
| | - Stephanie M Engel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Anita Haugan
- Norwegian Institute of Public Health, Oslo, Norway
| | | | - Gulnara Svyatova
- Scientific Center of Obstetrics, Gynecology and Perinatology, Almaty, Kazakhstan
| | - Nodira Zakhidova
- Institute of Immunology, Uzbek Academy of Sciences, Tashkent, Uzbekistan
| | - Dilbar Najmutdinova
- Republic Specialized Scientific Practical Medical Centre of Obstetrics and Gynecology, Tashkent, Uzbekistan
| | - Anna F Dominiczak
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Håkon K Gjessing
- Norwegian Institute of Public Health, Oslo, Norway
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Juan P Casas
- Farr Institute of Health Informatics, University College London, London, UK
| | - Frank Dudbridge
- Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - James J Walker
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Leeds, UK
| | | | - Unnur Thorsteinsdottir
- deCODE Genetics/Amgen, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Reynir T Geirsson
- Department of Obstetrics and Gynecology, Landspitali University Hospital, Reykjavik, Iceland
| | - Debbie A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Ann-Charlotte Iversen
- Centre of Molecular Inflammation Research (CEMIR) and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Per Magnus
- Norwegian Institute of Public Health, Oslo, Norway
| | - Hannele Laivuori
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kari Stefansson
- deCODE Genetics/Amgen, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Linda Morgan
- School of Life Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
49
|
Embryonic vascular disruption adverse outcomes: Linking high throughput signaling signatures with functional consequences. Reprod Toxicol 2017; 71:16-31. [DOI: 10.1016/j.reprotox.2017.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 04/03/2017] [Accepted: 04/07/2017] [Indexed: 11/23/2022]
|
50
|
Keshavarzi F, Mohammadpour-Gharehbagh A, Shahrakipour M, Teimoori B, Yazdi A, Yaghmaei M, Naroeei-Nejad M, Salimi S. The placental vascular endothelial growth factor polymorphisms and preeclampsia/preeclampsia severity. Clin Exp Hypertens 2017; 39:606-611. [PMID: 28665739 DOI: 10.1080/10641963.2017.1299751] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Preeclampsia (PE) is a serious pregnancy-specific condition, which originates from placenta and finishes after delivery. The present study has investigated the association between placental VEGF I/D (rs35569394), -1154G/A (rs1570360), and -634G/C(rs2010963) polymorphisms and maternal VEGF -2549 I/D (rs35569394) polymorphism with PE and PE severity. In this case-control study, the maternal blood of 217 women with PE and 210 normotensive pregnant women and the placenta of 84 PE women and 103 normotensive women were collected after delivery. Genotyping was done by PCR or PCR-RFLP methods. The maternal VEGF-2549I/D genotypes were not associated with PE or PE severity. The placental VEGF -2549 I/D genotypes were not associated with PE too; however; the placental VEGF-2549 DD genotype was statistically different between women with severe PE and mild PE or the controls. The placental VEGF -634GC and CC genotypes were significantly higher in PE women and associated with 2.6 and 2-fold higher risk of PE, respectively. The VEGF -634GC and CC genotypes were associated with PE severity. No association was found between placental VEGF -1154G/A polymorphism and PE or PE severity. The placental DGC haplotype of VEGF -2549 I/D, -1154G/A, and -634G/C polymorphisms was associated with 2.9-fold higher risk of PE. However, the placental IAG haplotype was associated with 0.3-fold lower risk of PE. In conclusion, the placental VEGF -2549 DD genotype was associated with severe PE and the placental -634GC and CC genotypes were associated with PE and severe PE. No association was found between VEGF -1154G/A polymorphism and PE or PE severity.
Collapse
Affiliation(s)
- Farshid Keshavarzi
- a Cellular and Molecular Research Center , Zahedan University of Medical Sciences , Zahedan , Iran.,b Department of Clinical Biochemistry, School of Medicine , Zahedan University of Medical Sciences , Zahedan , Iran
| | - Abbas Mohammadpour-Gharehbagh
- a Cellular and Molecular Research Center , Zahedan University of Medical Sciences , Zahedan , Iran.,b Department of Clinical Biochemistry, School of Medicine , Zahedan University of Medical Sciences , Zahedan , Iran
| | - Mahnaz Shahrakipour
- c Department of Biostatistics and Epidemiology, School of Public Health , Zahedan University of Medical Sciences , Zahedan , Iran
| | - Batool Teimoori
- d Department of Obstetrics and Gynecology, School of Medicine , Zahedan University of Medical Sciences , Zahedan , Iran.,e Pregnancy Health Research Center , Zahedan University of Medical Sciences , Zahedan , Iran
| | - Atefeh Yazdi
- d Department of Obstetrics and Gynecology, School of Medicine , Zahedan University of Medical Sciences , Zahedan , Iran.,e Pregnancy Health Research Center , Zahedan University of Medical Sciences , Zahedan , Iran
| | - Minoo Yaghmaei
- f Department of Obstetrics and Gynecology, School of Medicine , Shahid Beheshty University of Medical Sciences , Tehran , Iran
| | - Mehrnaz Naroeei-Nejad
- g Department of Genetics, School of Medicine , Zahedan University of Medical Sciences , Zahedan , Iran
| | - Saeedeh Salimi
- a Cellular and Molecular Research Center , Zahedan University of Medical Sciences , Zahedan , Iran.,b Department of Clinical Biochemistry, School of Medicine , Zahedan University of Medical Sciences , Zahedan , Iran
| |
Collapse
|