1
|
Elgazzaz M, Brawley A, Moronge D, Faulkner JL. Emerging Role of Leptin in Vascular and Placental Dysfunction in Preeclampsia. Arterioscler Thromb Vasc Biol 2025; 45:585-599. [PMID: 40177777 PMCID: PMC12036005 DOI: 10.1161/atvbaha.124.321676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Leptin is a well-known metabolic hormone that plays diverse roles in various body functions, including growth, reproduction, and blood pressure regulation. In pregnancy, leptin produced from the placenta is crucial for ensuring proper fetal development and angiogenesis; however, pathological increases in leptin in maternal circulation are strongly associated with vascular endothelial dysfunction and preeclampsia. Leptin has a strong role in fertility and healthy pregnancy; however, numerous clinical reports over the last 2 decades show that leptin levels pathologically increase in patients with preeclampsia independent of metabolic status (ie, obesity). Despite this strong correlation, the role of leptin in preeclampsia is largely unexplored compared with other biomarkers likely due to differences in placental leptin production among mammals. Emerging literature has recently begun to shed light on this hormone in preeclampsia pathogenesis and uncovered some key mechanisms whereby pathologically elevated leptin production leads to cardiovascular complications for pregnant women.
Collapse
Affiliation(s)
- Mona Elgazzaz
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Amalia Brawley
- Department of Obstetrics and Gynecology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Desmond Moronge
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Jessica L Faulkner
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Department of Obstetrics and Gynecology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| |
Collapse
|
2
|
Yu T, Wang G, Xu X, Yan J. Identification and validation of key biomarkers associated with immune and oxidative stress for preeclampsia by WGCNA and machine learning. Front Genet 2025; 16:1500061. [PMID: 40151199 PMCID: PMC11949101 DOI: 10.3389/fgene.2025.1500061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
Background Preeclampsia (PE), a major obstetric disorder marked by dysfunction in both placental and maternal vascular systems, continues to pose critical challenges in global maternal healthcare. This multisystem pregnancy complication contributes significantly to adverse perinatal outcomes and remains a leading cause of pregnancy-related morbidity worldwide. However, the available treatment options at present remain restricted. Our investigation employs an integrative bioinformatics approach to elucidate critical molecular signatures linked to the interplay between immunological dysregulation and oxidative stress mechanisms in PE pathogenesis. Methods In this study, we sourced the dataset from the GEO database with the aim of pinpointing differentially expressed genes (DEGs) between PE samples and control samples. Genes associated with oxidative stress were procured from the Genecards database. Next, we employed a comprehensive approach. This involved integrating WGCNA, GO and KEGG pathway analyses, constructing PPI networks, applying machine learning algorithms, performing gene GSEA, and conducting immune infiltration analysis to identify the key hub genes related to oxidative stress. Diagnostic potential of candidate biomarkers was quantitatively assessed through ROC curve modeling. Additionally, we constructed a miRNA - gene regulatory network for the identified diagnostic genes and predicted potential candidate drugs. In the final step, we validated the significant hub gene using independent external datasets, the hypoxia model of the HTR-8/SVneo cell line, and human placental tissue samples. Results At last, leptin (LEP) was identified as a core gene through screening and was found to be upregulated. The results of quantitative real-time polymerase chain reaction (qRT -PCR) and immunohistochemistry validation were consistent with those obtained from the datasets. KEGG analysis revealed that LEP was significantly enriched in "allograft rejection," "antigen processing," "ECM receptor interaction" and "graft versus host disease." GO analysis revealed that LEP was involved in biological processes such as "antigen processing and presentation," "peptide antigen assembly with MHC protein complex," "complex of collagen trimers," "MHC class II protein complex" and "mitochondrial protein containing complex." Moreover, immune cell analysis indicated that T follicular helper cells, plasmacytoid dendritic cells, neutrophils, and activated dendritic cells were positively correlated with LEP expression, whereas γδT cells, eosinophils, and central memory CD4+ T cells showed a negative correlation. These findings suggest that LEP influences the immune microenvironment of PE through its interaction with arious immune cells. In addition, 28 miRNAs and 15 drugs were predicted to target LEP. Finally, the overexpression of LEP was verified using independent external datasets, the hypoxia model of the HTR-8/SVneo cell line, and human placental tissue. Conclusion Through an integrated analytical framework employing WGCNA coupled with three distinct machine learning-driven phenotypic classification models, we discovered a pivotal regulatory gene. This gene has the potential to act as a novel diagnostic biomarker for PE. Moreover, it can be considered as a promising target for drug development related to PE. Notably, it shows a strong correlation with the immune microenvironment, suggesting its crucial role in the complex pathophysiological processes underlying PE.
Collapse
Affiliation(s)
- Tiantian Yu
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
- Fujian Clinical Research Center for Maternal - Fetal Medicine, Fuzhou, Fujian, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, Fujian, China
| | - Guiying Wang
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
- Fujian Clinical Research Center for Maternal - Fetal Medicine, Fuzhou, Fujian, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, Fujian, China
| | - Xia Xu
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
- Fujian Clinical Research Center for Maternal - Fetal Medicine, Fuzhou, Fujian, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, Fujian, China
| | - Jianying Yan
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
- Fujian Clinical Research Center for Maternal - Fetal Medicine, Fuzhou, Fujian, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, Fujian, China
| |
Collapse
|
3
|
Qi G, Gong Y, Li Y, Jin Y, Chi S, Zhang W, Luo X. Insufficient expression of COL6A1 promotes the development of early-onset severe preeclampsia by inhibiting the APJ/AKT signaling pathway. Cell Death Discov 2025; 11:81. [PMID: 40025063 PMCID: PMC11873267 DOI: 10.1038/s41420-025-02373-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/03/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025] Open
Abstract
Early-onset severe preeclampsia (eosPE) is one of the most severe complications of pregnancy. To identify the genes related to the development of eosPE. We downloaded and integrated analyzed microarray data from GSE44711, GSE66273, and GSE74341, which contains the expression profile of placental tissues from patients with eosPE and healthy controls. Our analysis revealed that collagen type VI alpha 1 (COL6A1) was downregulated in the eosPE placenta compared to normal pregnancy. COL6A1 promoted the migration, invasion and tube formation ability of HTR8/SVneo cells, HUVECs and primary extravillous trophoblasts (EVTs). To explore the underlying mechanisms, we conducted transcriptome sequencing, which indicated that the Apelin/APJ signaling pathway was affected by COL6A1 knockdown. In addition, we found that APJ expression was lower in the placental tissue of patients with eosPE compared to healthy pregnancies. Inhibition of APJ suppressed the invasion, migration, and tube formation abilities of trophoblasts. We also observed that COL6A1 increased the levels of p-AKT and p-mTOR, while the APJ inhibitor ML221 impaired this effect. Furthermore, transwell and tube formation assays demonstrated that ML221 attenuated the capabilities enhanced by COL6A1, an effect that could be rescued by the AKT activator SC79. Overall, these findings indicate that insufficient expression of COL6A1 attenuates the migration, invasion, and endothelial-like tube formation of HTR8/SVneo cells and primary EVTs via the APJ/AKT/mTOR pathway, thereby promoting the development of eosPE.
Collapse
Affiliation(s)
- Gonghua Qi
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital, Shandong University, Jinan, China
| | - Yanmin Gong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
| | - Yi Li
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
| | - Yanhui Jin
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
| | - Shuqi Chi
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
| | - Wenxia Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
| | - Xia Luo
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China.
| |
Collapse
|
4
|
Li Q, Wei X, Wu F, Qin C, Dong J, Chen C, Lin Y. Development and validation of preeclampsia predictive models using key genes from bioinformatics and machine learning approaches. Front Immunol 2024; 15:1416297. [PMID: 39544937 PMCID: PMC11560445 DOI: 10.3389/fimmu.2024.1416297] [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: 07/09/2024] [Accepted: 09/27/2024] [Indexed: 11/17/2024] Open
Abstract
Background Preeclampsia (PE) poses significant diagnostic and therapeutic challenges. This study aims to identify novel genes for potential diagnostic and therapeutic targets, illuminating the immune mechanisms involved. Methods Three GEO datasets were analyzed, merging two for training set, and using the third for external validation. Intersection analysis of differentially expressed genes (DEGs) and WGCNA highlighted candidate genes. These were further refined through LASSO, SVM-RFE, and RF algorithms to identify diagnostic hub genes. Diagnostic efficacy was assessed using ROC curves. A predictive nomogram and fully Connected Neural Network (FCNN) were developed for PE prediction. ssGSEA and correlation analysis were employed to investigate the immune landscape. Further validation was provided by qRT-PCR on human placental samples. Result Five biomarkers were identified with validation AUCs: CGB5 (0.663, 95% CI: 0.577-0.750), LEP (0.850, 95% CI: 0.792-0.908), LRRC1 (0.797, 95% CI: 0.728-0.867), PAPPA2 (0.839, 95% CI: 0.775-0.902), and SLC20A1 (0.811, 95% CI: 0.742-0.880), all of which are involved in key biological processes. The nomogram showed strong predictive power (C-index 0.873), while FCNN achieved an optimal AUC of 0.911 (95% CI: 0.732-1.000) in five-fold cross-validation. Immune infiltration analysis revealed the importance of T cell subsets, neutrophils, and NK cells in PE, linking these genes to immune mechanisms underlying PE pathogenesis. Conclusion CGB5, LEP, LRRC1, PAPPA2, and SLC20A1 are validated as key diagnostic biomarkers for PE. Nomogram and FCNN could credibly predict PE. Their association with immune infiltration underscores the crucial role of immune responses in PE pathogenesis.
Collapse
Affiliation(s)
- Qian Li
- Reproductive Medicine Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaowei Wei
- Reproductive Medicine Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Wu
- The International Peace Maternity and Child Health Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuanmei Qin
- Reproductive Medicine Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junpeng Dong
- Reproductive Medicine Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cailian Chen
- Department of Automation, Shanghai Jiao Tong University, Key Laboratory of System Control and Information Processing, Ministry of Education of China, Shanghai, China
| | - Yi Lin
- Reproductive Medicine Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Niclou AM, Cabre HE, Flanagan EW, Redman LM. Precision Interventions Targeting the Maternal Metabolic Milieu for Healthy Pregnancies in Obesity. Curr Diab Rep 2024; 24:227-235. [PMID: 39162956 PMCID: PMC12036336 DOI: 10.1007/s11892-024-01550-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/29/2024] [Indexed: 08/21/2024]
Abstract
PURPOSE OF REVIEW Entering pregnancy with obesity increases the risk of adverse health outcomes for parent and child. As such, research interventions are largely focused on limiting excess gestational weight gain during pregnancy, especially in those with obesity. Yet, while many lifestyle interventions are successful in reducing GWG, few affect pregnancy outcomes. Here we review work targeting the metabolic milieu instead of focusing solely on weight. RECENT FINDINGS Work done in non-pregnant populations suggests that specifically targeting glucose, triglyceride, and leptin levels or inflammatory makers improves the metabolic milieu and overall health. We posit that precision interventions that include strategies such as time restricted eating, following the 24 h movement guidelines, or reducing sedentary behavior during pregnancy can be successful approaches benefiting the maternal metabolic milieu and minimize the risk of adverse pregnancy outcomes. Personalized tools such as continuous glucose monitors or community-based approaches play an important role in pre-conception health and should be extrapolated to pregnancy interventions to directly benefit the metabolic milieu optimizing health outcomes for both parent and child.
Collapse
Affiliation(s)
- Alexandra M Niclou
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Hannah E Cabre
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Emily W Flanagan
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Leanne M Redman
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA.
| |
Collapse
|
6
|
Singh H, Almabhouh FA, Alshaikhli HSI, Hassan MJM, Daud S, Othman R, Md Salleh MFRR. Leptin in reproduction and hypertension in pregnancy. Reprod Fertil Dev 2024; 36:RD24060. [PMID: 39038160 DOI: 10.1071/rd24060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/27/2024] [Indexed: 07/24/2024] Open
Abstract
Leptin has important roles in numerous physiological functions, including those in the regulation of energy balance, and in immune and reproductive systems. However, in the recent years, evidence has implicated it in a number of obesity-related diseases, where its concentrations in serum are significantly elevated. Elevated serum leptin concentrations and increased placental leptin secretion have been reported in women with hypertensive disorders of pregnancy. Whether leptin is responsible for this disorder remains to be established. Leptin injections in healthy rats and mice during pregnancy result in endothelial activation, increased blood pressure and proteinuria. A potential role for leptin in the pathogenesis of pre-eclampsia is hypothesised, particularly in women who are overweight or obese where serum leptin concentrations are often elevated. This review summarises pertinent information in the literature on the role of leptin in puberty, pregnancy, and hypertensive disorders of pregnancy. In particular, the possible mechanism that may be involved in leptin-induced increase in blood pressure and proteinuria during pregnancy and the potential role of marinobufagenin in this disease entity. We hypothesise a significant role for oxidative stress in this, and propose a conceptual framework on the events that lead to endothelial activation, raised blood pressure and proteinuria following leptin administration.
Collapse
Affiliation(s)
- Harbindarjeet Singh
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Sg Buloh, Selangor, Malaysia
| | - Fayez A Almabhouh
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Sg Buloh, Selangor, Malaysia; and Department of Biology and Biotechnology, Faculty of Science Islamic University of Gaza, Gaza Strip, Palestine
| | | | | | - Suzanna Daud
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Sg Buloh, Selangor, Malaysia
| | - Rosfayati Othman
- Department of Physiology, Faculty of Medicine, MAHSA University, Bandar Saujana Putra, Kuala Langat, Selangor, Malaysia
| | - Muhd Fakh Rur Razi Md Salleh
- Department of Physiology, Faculty of Medicine, MAHSA University, Bandar Saujana Putra, Kuala Langat, Selangor, Malaysia
| |
Collapse
|
7
|
Schofield LG, Delforce SJ, Pryor JC, Endacott SK, Lumbers ER, Marshall SA, Pringle KG. The soluble (pro)renin receptor promotes a preeclampsia-like phenotype both in vitro and in vivo. Hypertens Res 2024; 47:1627-1641. [PMID: 38605139 PMCID: PMC11150152 DOI: 10.1038/s41440-024-01678-8] [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: 03/14/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024]
Abstract
Preeclampsia is classified as new-onset hypertension coupled with gross endothelial dysfunction. Placental (pro)renin receptor ((P)RR) and plasma soluble (P)RR (s(P)RR) are elevated in patients with preeclampsia. Thus, we aimed to interrogate the role (P)RR may play in the pathogenesis of preeclampsia. Human uterine microvascular endothelial cells (HUtMECs, n = 4) were cultured with either; vehicle (PBS), 25-100 nM recombinant s(P)RR, or 10 ng/ml TNF-a (positive control) for 24 h. Conditioned media and cells were assessed for endothelial dysfunction markers via qPCR, ELISA, and immunoblot. Angiogenic capacity was assessed through tube formation and adhesion assays. Additionally, pregnant rats were injected with an adenovirus overexpressing s(P)RR from mid-pregnancy (day 8.5), until term (n = 6-7 dams/treatment). Maternal and fetal tissues were assessed. HUtMECs treated with recombinant s(P)RR displayed increased expression of endothelial dysfunction makers including vascular cell adhesion molecule-1, intracellular adhesion molecule-1, and endothelin-1 mRNA expression (P = 0.003, P = 0.001, P = 0.009, respectively), along with elevated endothelin-1 protein secretion (P < 0.001) compared with controls. Recombinant s(P)RR impaired angiogenic capacity decreasing the number of branches, total branch length, and mesh area (P < 0.001, P = 0.004, and P = 0.009, respectively), while also increasing vascular adhesion (P = 0.032). +ADV rats exhibited increased systolic (P = 0.001), diastolic (P = 0.010), and mean arterial pressures (P = 0.012), compared with -ADV pregnancies. Renal arteries from +ADV-treated rats had decreased sensitivity to acetylcholine-induced relaxation (P = 0.030), compared with -ADV pregnancies. Our data show that treatment with s(P)RR caused hypertension and growth restriction in vivo and caused marked endothelial dysfunction in vitro. These findings demonstrate the significant adverse actions of s(P)RR on vascular dysfunction that is characteristic of the preeclamptic phenotype.
Collapse
Affiliation(s)
- Lachlan G Schofield
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Sarah J Delforce
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Jennifer C Pryor
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Immune Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- National Health & Medical Research Council (NHMRC) Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia
| | - Saije K Endacott
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Eugenie R Lumbers
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Sarah A Marshall
- Department of Obstetrics and Gynaecology, The Ritchie Centre, School of Clinical Sciences, Monash University and The Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Kirsty G Pringle
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia.
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia.
| |
Collapse
|
8
|
Marić I, Stevenson DK, Aghaeepour N, Gaudillière B, Wong RJ, Angst MS. Predicting Preterm Birth Using Proteomics. Clin Perinatol 2024; 51:391-409. [PMID: 38705648 PMCID: PMC11186213 DOI: 10.1016/j.clp.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
The complexity of preterm birth (PTB), both spontaneous and medically indicated, and its various etiologies and associated risk factors pose a significant challenge for developing tools to accurately predict risk. This review focuses on the discovery of proteomics signatures that might be useful for predicting spontaneous PTB or preeclampsia, which often results in PTB. We describe methods for proteomics analyses, proteomics biomarker candidates that have so far been identified, obstacles for discovering biomarkers that are sufficiently accurate for clinical use, and the derivation of composite signatures including clinical parameters to increase predictive power.
Collapse
Affiliation(s)
- Ivana Marić
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, 453 Quarry Road, Palo Alto, CA 94304, USA.
| | - David K Stevenson
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, 453 Quarry Road, Palo Alto, CA 94304, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Grant Building, Office 276A, 300 Pasteur Drive, Stanford, CA 94305-5117, USA; Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Grant S280, Stanford, CA 94305, USA
| | - Brice Gaudillière
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Grant Building, Office 276A, 300 Pasteur Drive, Stanford, CA 94305-5117, USA; Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Grant S280, Stanford, CA 94305, USA
| | - Ronald J Wong
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, 453 Quarry Road, Palo Alto, CA 94304, USA
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Grant Building, Office 276A, 300 Pasteur Drive, Stanford, CA 94305-5117, USA
| |
Collapse
|
9
|
Rumer KK, Sehgal S, Kramer A, Bogart KP, Winn VD. The effects of leptin on human cytotrophoblast invasion are gestational age and dose-dependent. Front Endocrinol (Lausanne) 2024; 15:1386309. [PMID: 38846494 PMCID: PMC11154010 DOI: 10.3389/fendo.2024.1386309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/08/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Leptin and its receptors are expressed by the human placenta throughout gestation, yet the role of leptin in early human placental development is not well characterized. Leptin is overexpressed in the placentas from preeclamptic (PE) pregnancies. PE can result from the impaired invasion of fetal placental cells, cytotrophoblasts (CTBs), into the maternal decidua. We hypothesized that elevated leptin levels would impair human CTB invasion. Methods The effects of leptin on the invasion of human CTBs were evaluated in three cell models, HTR-8/SVneo cells, primary CTBs, and placental villous explants using invasion assays. Further, leptin receptor expression was characterized in all three cell models using RT-PCR. Further phosphokinase assays were performed in HTR-8/SVneo cells to determine signaling pathways involved in CTB invasion in response to differential leptin doses. Results We found that, prior to 8 weeks gestation, leptin promoted CTB invasion in the explant model. After 11 weeks gestation in explants, primary CTBs and in HTR-8/SVneo cells, leptin promoted invasion at moderate but not at high concentrations. Further, leptin receptor characterization revealed that leptin receptor expression did not vary over gestation, however, STAT, PI3K and MAPK pathways showed different signaling in response to varied leptin doses. Discussion These data suggest that the excess placental leptin observed in PE may cause impaired CTB invasion as a second-trimester defect. Leptin's differential effect on trophoblast invasion may explain the role of hyperleptinemia in preeclampsia pathogenesis.
Collapse
Affiliation(s)
- Kristen K. Rumer
- Department of Obstetrics and Gynecology, University of Colorado, Aurora, CO, United States
- Department of Molecular and Cellular Medicine, University of Colorado, Aurora, CO, United States
| | - Shilpi Sehgal
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Anita Kramer
- Department of Obstetrics and Gynecology, University of Colorado, Aurora, CO, United States
| | - Kevin P. Bogart
- Department of Obstetrics and Gynecology, University of Colorado, Aurora, CO, United States
| | - Virginia D. Winn
- Department of Obstetrics and Gynecology, University of Colorado, Aurora, CO, United States
- Department of Molecular and Cellular Medicine, University of Colorado, Aurora, CO, United States
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
10
|
Taliento C, Piccolotti I, Sabattini A, Tormen M, Cappadona R, Greco P, Scutiero G. Effect of Physical Activity during Pregnancy on the Risk of Hypertension Disorders and Gestational Diabetes: Evidence Generated by New RCTs and Systematic Reviews. J Clin Med 2024; 13:2198. [PMID: 38673471 PMCID: PMC11050519 DOI: 10.3390/jcm13082198] [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: 02/06/2024] [Revised: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Hypertensive disorders of pregnancy (HDP) and gestational diabetes mellitus (GDM) are the most common medical complications in pregnancy. Physical exercise is considered to be safe and beneficial during pregnancy. Moreover, pregnancy could be considered as an opportunity for healthcare providers to promote positive lifestyle behavior and optimize the well-being of pregnant women. Since there are few up-to-date reviews evaluating the role of exercise and the risks of developing obstetrical complications, we performed a review to investigate the effects of physical activity and exercise during pregnancy compared to a control group, focusing on the risk of development of HDP and GDM. We searched Medline and Web of Science, including only randomized controlled trials (RCTs) and systematic reviews. This review supports a beneficial effect of exercise and provides evidence that it significantly decreases the risk of HDP and GDM.
Collapse
Affiliation(s)
- Cristina Taliento
- Department of Medical Sciences, Obstetrics and Gynecology Unit, University Hospital “Sant’Anna”, 44121 Ferrara, Italy; (C.T.); (I.P.); (A.S.); (M.T.); (R.C.); (G.S.)
- Department of Development and Regeneration—Woman and Child, KU Leuven, 3000 Leuven, Belgium
| | - Irene Piccolotti
- Department of Medical Sciences, Obstetrics and Gynecology Unit, University Hospital “Sant’Anna”, 44121 Ferrara, Italy; (C.T.); (I.P.); (A.S.); (M.T.); (R.C.); (G.S.)
| | - Arianna Sabattini
- Department of Medical Sciences, Obstetrics and Gynecology Unit, University Hospital “Sant’Anna”, 44121 Ferrara, Italy; (C.T.); (I.P.); (A.S.); (M.T.); (R.C.); (G.S.)
| | - Mara Tormen
- Department of Medical Sciences, Obstetrics and Gynecology Unit, University Hospital “Sant’Anna”, 44121 Ferrara, Italy; (C.T.); (I.P.); (A.S.); (M.T.); (R.C.); (G.S.)
| | - Rosaria Cappadona
- Department of Medical Sciences, Obstetrics and Gynecology Unit, University Hospital “Sant’Anna”, 44121 Ferrara, Italy; (C.T.); (I.P.); (A.S.); (M.T.); (R.C.); (G.S.)
| | - Pantaleo Greco
- Department of Medical Sciences, Obstetrics and Gynecology Unit, University Hospital “Sant’Anna”, 44121 Ferrara, Italy; (C.T.); (I.P.); (A.S.); (M.T.); (R.C.); (G.S.)
- Department of Medical Sciences, Institute of Obstetrics and Gynecology, University of Ferrara, Via Fossato di Mortara, 64/B, 44121 Ferrara, Italy
| | - Gennaro Scutiero
- Department of Medical Sciences, Obstetrics and Gynecology Unit, University Hospital “Sant’Anna”, 44121 Ferrara, Italy; (C.T.); (I.P.); (A.S.); (M.T.); (R.C.); (G.S.)
| |
Collapse
|
11
|
Adibi JJ, Zhao Y, Koistinen H, Mitchell RT, Barrett ES, Miller R, O'Connor TG, Xun X, Liang HW, Birru R, Smith M, Moog NK. Molecular pathways in placental-fetal development and disruption. Mol Cell Endocrinol 2024; 581:112075. [PMID: 37852527 PMCID: PMC10958409 DOI: 10.1016/j.mce.2023.112075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/11/2023] [Accepted: 09/24/2023] [Indexed: 10/20/2023]
Abstract
The first trimester of pregnancy ranks high in priority when minimizing harmful exposures, given the wide-ranging types of organogenesis occurring between 4- and 12-weeks' gestation. One way to quantify potential harm to the fetus in the first trimester is to measure a corollary effect on the placenta. Placental biomarkers are widely present in maternal circulation, cord blood, and placental tissue biopsied at birth or at the time of pregnancy termination. Here we evaluate ten diverse pathways involving molecules expressed in the first trimester human placenta based on their relevance to normal fetal development and to the hypothesis of placental-fetal endocrine disruption (perturbation in development that results in abnormal endocrine function in the offspring), namely: human chorionic gonadotropin (hCG), thyroid hormone regulation, peroxisome proliferator activated receptor protein gamma (PPARγ), leptin, transforming growth factor beta, epiregulin, growth differentiation factor 15, small nucleolar RNAs, serotonin, and vitamin D. Some of these are well-established as biomarkers of placental-fetal endocrine disruption, while others are not well studied and were selected based on discovery analyses of the placental transcriptome. A literature search on these biomarkers summarizes evidence of placenta-specific production and regulation of each biomarker, and their role in fetal reproductive tract, brain, and other specific domains of fetal development. In this review, we extend the theory of fetal programming to placental-fetal programming.
Collapse
Affiliation(s)
- Jennifer J Adibi
- Department of Epidemiology, University of Pittsburgh School of Public Health, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Yaqi Zhao
- St. Jude's Research Hospital, Memphis, TN, USA
| | - Hannu Koistinen
- Department of Clinical Chemistry, University of Helsinki, Helsinki, Finland
| | - Rod T Mitchell
- Department of Paediatric Endocrinology, Royal Hospital for Children and Young People, Edinburgh BioQuarter, Edinburgh, UK
| | - Emily S Barrett
- Environmental and Population Health Bio-Sciences, Rutgers University School of Public Health, Piscataway, NJ, USA
| | - Richard Miller
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA
| | - Thomas G O'Connor
- Department of Psychiatry, University of Rochester Medical Center, Rochester, NY, USA
| | - Xiaoshuang Xun
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Hai-Wei Liang
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Rahel Birru
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Megan Smith
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nora K Moog
- Department of Medical Psychology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
12
|
Ozmen A, Nwabuobi C, Tang Z, Guo X, Larsen K, Guller S, Blas J, Moore M, Kayisli UA, Lockwood CJ, Guzeloglu-Kayisli O. Leptin-Mediated Induction of IL-6 Expression in Hofbauer Cells Contributes to Preeclampsia Pathogenesis. Int J Mol Sci 2023; 25:135. [PMID: 38203306 PMCID: PMC10778808 DOI: 10.3390/ijms25010135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Leptin plays a crucial role in regulating energy homoeostasis, neuroendocrine function, metabolism, and immune and inflammatory responses. The adipose tissue is a main source of leptin, but during pregnancy, leptin is also secreted primarily by the placenta. Circulating leptin levels peak during the second trimester of human pregnancy and fall after labor. Several studies indicated a strong association between elevated placental leptin levels and preeclampsia (PE) pathogenesis and elevated serum interleukin-6 (IL-6) levels in PE patients. Therefore, we hypothesized that a local increase in placental leptin production induces IL-6 production in Hofbauer cells (HBCs) to contribute to PE-associated inflammation. We first investigated HBCs-specific IL-6 and leptin receptor (LEPR) expression and compared their immunoreactivity in PE vs. gestational age-matched control placentas. Subsequently, we examined the in vitro regulation of IL-6 as well as the phosphorylation levels of intracellular signaling proteins STAT3, STAT5, NF-κB, and ERK1/2 by increasing recombinant human leptin concentrations (10 to 1000 ng/mL) in primary cultured HBCs. Lastly, HBC cultures were incubated with leptin ± specific inhibitors of STAT3 or STAT5, or p65 NF-κB or ERK1/2 MAPK signaling cascades to determine relevant cascade(s) involved in leptin-mediated IL-6 regulation. Immunohistochemistry revealed ~three- and ~five-fold increases in IL-6 and LEPR expression, respectively, in HBCs from PE placentas. In vitro analysis indicated that leptin treatment in HBCs stimulate IL-6 in a concentration-dependent manner both at the transcriptional and secretory levels (p < 0.05). Moreover, leptin-treated HBC cultures displayed significantly increased phosphorylation levels of STAT5, p65 NF-κB, and ERK1/2 MAPK and pre-incubation of HBCs with a specific ERK1/2 MAPK inhibitor blocked leptin-induced IL-6 expression. Our in situ results show that HBCs contribute to the pathogenesis of PE by elevating IL-6 expression, and in vitro results indicate that induction of IL-6 expression in HBCs is primarily leptin-mediated. While HBCs display an anti-inflammatory phenotype in normal placentas, elevated levels of leptin may transform HBCs into a pro-inflammatory phenotype by activating ERK1/2 MAPK to augment IL-6 expression.
Collapse
Affiliation(s)
- Asli Ozmen
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| | - Chinedu Nwabuobi
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| | - Zhonghua Tang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA; (Z.T.); (S.G.)
| | - Xiaofang Guo
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| | - Kellie Larsen
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| | - Seth Guller
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA; (Z.T.); (S.G.)
| | - Jacqueline Blas
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| | - Monica Moore
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| | - Umit A. Kayisli
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| | - Charles J. Lockwood
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| | - Ozlem Guzeloglu-Kayisli
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| |
Collapse
|
13
|
Awoyemi T, Zhang W, Rahbar M, Cribbs A, Logenthiran P, Jiang S, Collett G, Cerdeira AS, Vatish M. A cross-sectional analysis of syncytiotrophoblast membrane extracellular vesicles-derived transcriptomic biomarkers in early-onset preeclampsia. Front Cardiovasc Med 2023; 10:1291642. [PMID: 38099221 PMCID: PMC10720444 DOI: 10.3389/fcvm.2023.1291642] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023] Open
Abstract
Background Preeclampsia (PE) is a pregnancy-specific hypertensive disorder affecting 2%-8% of pregnancies worldwide. Biomarker(s) for the disorder exists, but while these have excellent negative predictive value, their positive predictive value is poor. Extracellular vesicles released by the placenta into the maternal circulation, syncytiotrophoblast membrane extracellular vesicles (STB-EVs), have been identified as being involved in PE with the potential to act as liquid biopsies. Objective The objective of this study was to identify the difference in the transcriptome of placenta and STB-EVs between preeclampsia and normal pregnancy (NP) and mechanistic pathways. Methods/study design We performed RNA-sequencing on placental tissue, medium/large and small STB-EVs from PE (n = 6) and NP (n = 6), followed by bioinformatic analysis to identify targets that could be used in the future for EV-based diagnostic tests for preeclampsia. Some of the identified biomarkers were validated with real-time polymerase chain reactions. Results Our analysis identified a difference in the transcriptomic STB-EV cargo between PE and NP. We then identified and verified the differential expression of FLNB, COL17A1, SLC45A4, LEP, HTRA4, PAPP-A2, EBI3, HSD17B1, FSTL3, INHBA, SIGLEC6, and CGB3. Our analysis also identified interesting mechanistic processes via an in silico prediction of STB-EV-based mechanistic pathways. Conclusions In this study, using comprehensive profiling of differentially expressed/carried genes of three linked sample subtypes in PE, we identified potential biomarkers and mechanistic gene pathways that may be important in the pathophysiology of PE and could be further explored in future studies.
Collapse
Affiliation(s)
- Toluwalase Awoyemi
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Wei Zhang
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Maryam Rahbar
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Adam Cribbs
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Prasanna Logenthiran
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Shuhan Jiang
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Gavin Collett
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Ana Sofia Cerdeira
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Manu Vatish
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
14
|
Zhou S, Li J, Yang W, Xue P, Yin Y, Wang Y, Tian P, Peng H, Jiang H, Xu W, Huang S, Zhang R, Wei F, Sun HX, Zhang J, Zhao L. Noninvasive preeclampsia prediction using plasma cell-free RNA signatures. Am J Obstet Gynecol 2023; 229:553.e1-553.e16. [PMID: 37211139 DOI: 10.1016/j.ajog.2023.05.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/02/2023] [Accepted: 05/14/2023] [Indexed: 05/23/2023]
Abstract
BACKGROUND Preeclampsia, especially preterm preeclampsia and early-onset preeclampsia, is a life-threating pregnancy disorder, and the heterogeneity and complexity of preeclampsia make it difficult to predict risk and to develop treatments. Plasma cell-free RNA carries unique information from human tissue and may be useful for noninvasive monitoring of maternal, placental, and fetal dynamics during pregnancy. OBJECTIVE This study aimed to investigate various RNA biotypes associated with preeclampsia in plasma and to develop classifiers to predict preterm preeclampsia and early-onset preeclampsia before diagnosis. STUDY DESIGN We performed a novel, cell-free RNA sequencing method termed polyadenylation ligation-mediated sequencing to investigate the cell-free RNA characteristics of 715 healthy pregnancies and 202 pregnancies affected by preeclampsia before symptom onset. We explored differences in the abundance of different RNA biotypes in plasma between healthy and preeclampsia samples and built preterm preeclampsia and early-onset preeclampsia prediction classifiers using machine learning methods. Furthermore, we validated the performance of the classifiers using the external and internal validation cohorts and assessed the area under the curve and positive predictive value. RESULTS We detected 77 genes, including messenger RNA (44%) and microRNA (26%), that were differentially expressed in healthy mothers and mothers with preterm preeclampsia before symptom onset, which could separate participants with preterm preeclampsia from healthy samples and that played critical functional roles in preeclampsia physiology. We developed 2 classifiers for predicting preterm preeclampsia and early-onset preeclampsia before diagnosis based on 13 cell-free RNA signatures and 2 clinical features (in vitro fertilization and mean arterial pressure), respectively. Notably, both classifiers showed enhanced performance when compared with the existing methods. The preterm preeclampsia prediction model achieved 81% area under the curve and 68% positive predictive value in an independent validation cohort (preterm, n=46; control, n=151); the early-onset preeclampsia prediction model had an area under the curve of 88% and a positive predictive value of 73% in an external validation cohort (early-onset preeclampsia, n=28; control, n=234). Furthermore, we demonstrated that downregulation of microRNAs may play vital roles in preeclampsia through the upregulation of preeclampsia-relevant target genes. CONCLUSION In this cohort study, a comprehensive transcriptomic landscape of different RNA biotypes in preeclampsia was presented and 2 advanced classifiers with substantial clinical importance for preterm preeclampsia and early-onset preeclampsia prediction before symptom onset were developed. We demonstrated that messenger RNA, microRNA, and long noncoding RNA can simultaneously serve as potential biomarkers of preeclampsia, holding the promise of prevention of preeclampsia in the future. Abnormal cell-free messenger RNA, microRNA, and long noncoding RNA molecular changes may help to elucidate the pathogenic determinants of preeclampsia and open new therapeutic windows to effectively reduce pregnancy complications and fetal morbidity.
Collapse
Affiliation(s)
- Si Zhou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China; BGI Genomics, BGI-Shenzhen, Shenzhen, China; Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Shijiazhuang BGI Genomics Co, Ltd, Shijiazhuang, Hebei Province, China; Shijiazhuang BGI Clinical Laboratory Co, Ltd, Shijiazhuang, Hebei Province, China
| | - Jie Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China; BGI-Shenzhen, Shenzhen, China; BGI-Beijing, Beijing, China
| | - Wenzhi Yang
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Shijiazhuang BGI Genomics Co, Ltd, Shijiazhuang, Hebei Province, China; Shijiazhuang BGI Clinical Laboratory Co, Ltd, Shijiazhuang, Hebei Province, China
| | - Penghao Xue
- Shijiazhuang BGI Clinical Laboratory Co, Ltd, Shijiazhuang, Hebei Province, China
| | - Yanning Yin
- Shijiazhuang BGI Clinical Laboratory Co, Ltd, Shijiazhuang, Hebei Province, China
| | - Yunfang Wang
- Shijiazhuang BGI Clinical Laboratory Co, Ltd, Shijiazhuang, Hebei Province, China
| | | | | | | | - Wenqiu Xu
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Shang Huang
- Shenzhen Children's Hospital of China Medical University, Shenzhen, China
| | - Rui Zhang
- Division of Maternal-Fetal Medicine, Jinan University-affiliated Shenzhen Baoan Women's and Children's Hospital, Shenzhen, China.
| | - Fengxiang Wei
- Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Shenzhen, China.
| | - Hai-Xi Sun
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China; BGI-Shenzhen, Shenzhen, China; BGI-Beijing, Beijing, China.
| | - Jianguo Zhang
- BGI Genomics, BGI-Shenzhen, Shenzhen, China; Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Shijiazhuang BGI Genomics Co, Ltd, Shijiazhuang, Hebei Province, China; Shijiazhuang BGI Clinical Laboratory Co, Ltd, Shijiazhuang, Hebei Province, China.
| | - Lijian Zhao
- BGI Genomics, BGI-Shenzhen, Shenzhen, China; Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Shijiazhuang BGI Genomics Co, Ltd, Shijiazhuang, Hebei Province, China; Medical Technology College of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
15
|
Enebe JT, Enebe NO, Nwagha TU, Meka IA, Nwankwo ME, Izuka EO, Egede JO, Ugwu IA, Okoro NI, Okoye HC, Iyoke CA. Serum leptin levels and relationship with maternal weight gain at term among obese and non-obese pregnant women in Enugu, Nigeria: a comparative cross-sectional study. J Int Med Res 2023; 51:3000605231213265. [PMID: 38017364 PMCID: PMC10686020 DOI: 10.1177/03000605231213265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 10/23/2023] [Indexed: 11/30/2023] Open
Abstract
OBJECTIVE To determine and compare the mean maternal serum leptin levels, the prevalence of high serum leptin levels and mean gestational weight gain at term among obese and non-obese pregnant women in Enugu, Nigeria. METHODS This cross-sectional comparative study enrolled obese and non-obese pregnant women. The serum leptin levels of the women were determined using an enzyme-linked immunosorbent assay kit. Anthropometric and sociodemographic data were obtained and compared. Mean weight gain during pregnancy was determined. RESULTS A total of 170 pregnant women were included in the study. The mean ± SD serum leptin level (99.39 ± 50.2 ng/ml) and the prevalence of hyperleptinaemia (81 of 85 patients; 95.3%) among the obese pregnant women at term were significantly higher than those of the non-obese pregnant women (48.98 ± 30.35 ng/ml/65 of 85 patients; 76.5%). The mean percentage weight gain was significantly higher in the non-obese women compared with the obese women at term. The predictors of high maternal serum leptin level at term among the participants were the employment status and levels of education of the participants. CONCLUSION Maternal serum leptin level, maternal weight gain and prevalence of hyperleptinaemia at term were significantly higher in the obese compared with the non-obese pregnant women.
Collapse
Affiliation(s)
- Joseph Tochukwu Enebe
- Department of Obstetrics and Gynaecology, Enugu State University of Science and Technology, College of Medicine/Teaching Hospital, Parklane, Enugu, Nigeria
| | - Nympha Onyinye Enebe
- Department of Community Medicine, University of Nigeria Teaching Hospital (UNTH), Ituku Ozalla, Enugu, Nigeria
| | - Theresa Ukamaka Nwagha
- Department of Haematology and Immunology, University of Nigeria Teaching Hospital (UNTH), Ituku Ozalla, Enugu, Nigeria
- Department of Haematology and Immunology, College of Medicine, Ituku Ozalla Campus, Enugu, Nigeria
| | - Ijeoma Angela Meka
- Department of Chemical Pathology, College of Medicine, University of Nigeria Teaching Hospital (UNTH), Ituku Ozalla, Enugu, Nigeria
| | - Malackay Ezenwaeze Nwankwo
- Department of Obstetrics and Gynaecology, Enugu State University of Science and Technology, College of Medicine/Teaching Hospital, Parklane, Enugu, Nigeria
| | - Emmanuel Obiora Izuka
- Department of Obstetrics and Gynaecology, University of Nigeria Teaching Hospital (UNTH), Ituku Ozalla, Enugu, Nigeria
| | - John Okafor Egede
- Department of Obstetrics and Gynaecology, Alex Ekwueme Federal University Teaching Hospital, Abakaliki, Nigeria
| | - Innocent Anayochukwu Ugwu
- Department of Obstetrics and Gynaecology, Enugu State University of Science and Technology, College of Medicine/Teaching Hospital, Parklane, Enugu, Nigeria
| | - Ngozi Ijeoma Okoro
- Department of Chemical Pathology, College of Medicine, Enugu State University of Science and Technology (ESUT), Enugu, Nigeria
| | - Helen Chioma Okoye
- Department of Haematology and Immunology, University of Nigeria Teaching Hospital (UNTH), Ituku Ozalla, Enugu, Nigeria
| | - Chukwuemeka Anthony Iyoke
- Department of Obstetrics and Gynaecology, College of Medicine, University of Nigeria Ituku/Ozalla Campus, Enugu State, Nigeria
| |
Collapse
|
16
|
Valencia-Ortega J, Solis-Paredes JM, Saucedo R, Estrada-Gutierrez G, Camacho-Arroyo I. Excessive Pregestational Weight and Maternal Obstetric Complications: The Role of Adipokines. Int J Mol Sci 2023; 24:14678. [PMID: 37834125 PMCID: PMC10572963 DOI: 10.3390/ijms241914678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
There is a high frequency of overweight and obesity in women of reproductive age. Women who start pregnancy with overweight or obesity have an increased risk of developing maternal obstetric complications such as gestational hypertension, pre-eclampsia, gestational diabetes mellitus, postpartum hemorrhage, and requiring C-section to resolve the pregnancy with a higher risk of C-section surgical site infection. Excessive weight in pregnancy is characterized by dysregulation of adipokines, the functions of which partly explain the predisposition of pregnant women with overweight or obesity to these maternal obstetric complications. This review compiles, organizes, and analyzes the most recent studies on adipokines in pregnant women with excess weight and the potential pathophysiological mechanisms favoring the development of maternal pregnancy complications.
Collapse
Affiliation(s)
- Jorge Valencia-Ortega
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 11000, Mexico;
| | - Juan Mario Solis-Paredes
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico;
| | - Renata Saucedo
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | | | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 11000, Mexico;
| |
Collapse
|
17
|
Deng J, Zhao HJ, Zhong Y, Hu C, Meng J, Wang C, Lan X, Wang X, Chen ZJ, Yan J, Wang W, Li Y. H3K27me3-modulated Hofbauer cell BMP2 signalling enhancement compensates for shallow trophoblast invasion in preeclampsia. EBioMedicine 2023; 93:104664. [PMID: 37331163 DOI: 10.1016/j.ebiom.2023.104664] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/20/2023] Open
Abstract
BACKGROUND Preeclampsia (PE) is a common hypertensive pregnancy disorder associated with shallow trophoblast invasion. Although bone morphogenetic protein 2 (BMP2) has been shown to promote trophoblast invasion in vitro, its cellular origin and molecular regulation in placenta, as well as its potential role in PE, has yet to be established. Additionally, whether BMP2 and/or its downstream molecules could serve as potential diagnostic or therapeutic targets for PE has not been explored. METHODS Placentas and sera from PE and healthy pregnant women were subjected to multi-omics analyses, immunoblots, qPCR, and ELISA assays. Immortalized trophoblast cells, primary cultures of human trophoblasts, and first-trimester villous explants were used for in vitro experiments. Adenovirus expressing sFlt-1 (Ad Flt1)-induced PE rat model was used for in vivo studies. FINDINGS We find globally decreased H3K27me3 modifications and increased BMP2 signalling in preeclamptic placentas, which is negatively correlated with clinical manifestations. BMP2 is derived from Hofbauer cells and epigenetically regulated by H3K27me3 modification. BMP2 promotes trophoblast invasion and vascular mimicry by upregulating BMP6 via BMPR1A-SMAD2/3-SMAD4 signalling. BMP2 supplementation alleviates high blood pressure and fetal growth restriction phenotypes in Ad Flt1-induced rat PE model. INTERPRETATION Our findings demonstrate that epigenetically regulated Hofbauer cell-derived BMP2 signalling enhancement in late gestation could serve as a compensatory response for shallow trophoblast invasion in PE, suggesting opportunities for diagnostic marker and therapeutic target applications in PE clinical management. FUNDING National Key Research and Development Program of China (2022YFC2702400), National Natural Science Foundation of China (82101784, 82171648, 31988101), and Natural Science Foundation of Shandong Province (ZR2020QH051, ZR2020MH039).
Collapse
Affiliation(s)
- Jianye Deng
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China; Medical Integration and Practice Center, Shandong University, Jinan, Shandong, 250012, China
| | - Hong-Jin Zhao
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Ying Zhong
- Cardiovascular Research Center of the General Medical Services, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - Cuiping Hu
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jinlai Meng
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Chunling Wang
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xiangxin Lan
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xiyao Wang
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Junhao Yan
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Wei Wang
- Division of Neonatology, Department of Pediatrics, Massachusetts General Hospital, Boston, MA 02115, USA.
| | - Yan Li
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China; Medical Integration and Practice Center, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
18
|
Wang Y, Bai X, Guo X, Gao X, Chen Y, Li H, Fan W, Han C. Bioinformatics analysis combined with clinical sample screening reveals that leptin may be a biomarker of preeclampsia. Front Physiol 2023; 13:1031950. [PMID: 36685185 PMCID: PMC9846503 DOI: 10.3389/fphys.2022.1031950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction: Preeclampsia (PE) is a gestational hypertensive disease with unclear pathogenesis. This study aimed to identify the genes that play an important role in determining the pathogenesis of PE using bioinformatics analysis and fundamental researches. Materials and methods: Datasets from the Gene Expression Omnibus (GEO) database were used to screen for differentially expressed genes (DEGs). The NCBI, SangerBox, and other databases were used to analyze the functions of the DEGs. Targetscan7, miRWalk, ENCORI, DIANA TOOLS, CircBank databases, and the Cytoscape tool were used to construct the lncRNA/circRNA-miRNA- LEP network. SRAMP, RPISeq, RBPsuite, and catRPAID were used to analyze the RNA modifications of LEP. Immune cell infiltration was analyzed using the dataset GSE75010. Placental tissues from normal pregnant women and PE patients were collected, screened for gene expression using reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blotting. The results were further verified in HTR-8/SVneo cell line hypoxia model and PE mouse model. Results: Our analyses revealed that LEP was significantly upregulated in eight datasets. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses indicated that LEP was involved in the JAK/STAT signaling pathway, angiogenesis, and placental development. Immune cell infiltration analysis showed that M1 and M2 macrophages differed between normal pregnancies and those in PE patients. A competing endogenous RNA (ceRNA) network was constructed, and proteins interacting with LEP were identified. RNA modification sites of LEP were also identified. Finally, the overexpression of LEP in PE was confirmed in clinical samples, HTR-8/SVneo cell line and PE mouse model. Conclusion: Our results indicate that LEP overexpression is associated with PE and may be a potential diagnostic marker and therapeutic target.
Collapse
Affiliation(s)
- Yajuan Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuening Bai
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Guo
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoli Gao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuanyuan Chen
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Huanrong Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenjun Fan
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Cha Han
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China,*Correspondence: Cha Han,
| |
Collapse
|
19
|
Okafor LU, Eleje GU, Ikechebelu JI, Ugwu EO, Nwosu OB, Ikpeze OC, Udigwe GO, Eke AC, Okoli SA, Oguejiofor CB, Ogabido CA, Malachy DE, Ofor IJ, Okafor CG. Maternal and umbilical cord blood levels of leptin in pre-eclamptic and healthy pregnant women: A comparative cross-sectional study. WOMEN'S HEALTH (LONDON, ENGLAND) 2023; 19:17455057231213272. [PMID: 38047453 PMCID: PMC10697037 DOI: 10.1177/17455057231213272] [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: 02/22/2023] [Revised: 09/27/2023] [Accepted: 10/24/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND Pre-eclampsia is a multi-systemic disease with its attendant increased maternal and perinatal morbidities and mortality. It has been hypothesized that leptin contributes immensely to the natural history of pre-eclampsia. However, there is considerable disagreement in the reports of existing research work on the link between fetomaternal serum leptin levels and pre-eclampsia. OBJECTIVE To determine and compare the maternal and umbilical cord sera levels of leptin in women with pre-eclampsia and healthy pregnant women. STUDY DESIGN This is an analytical cross-sectional study. METHODS The study involved consenting 120 pregnant participants (60 on each arm). Pregnant women diagnosed with pre-eclampsia constituted the investigation group, while the controls were normotensive pregnant women. They were matched for maternal age and body mass index. Venous blood specimens were obtained from the participants for assessment of the serum leptin concentration while umbilical cord blood samples were obtained following delivery of the neonate in advance of the removal of the placenta. The collected blood samples were analysed for the levels of leptin in a blinded pattern. The primary outcome measures were maternal serum leptin levels and umbilical cord serum leptin levels. RESULTS Mean maternal serum leptin concentration in the pre-eclampsia group was significantly higher than that in the control group (24.88 ± 3.92 vs. 15.03 ± 2.98ng/mL, p < 0.001). Similarly, maternal serum leptin concentration was significantly higher in participants with severe pre-eclampsia compared with those with mild pre-eclampsia (25.91 ± 3.5 vs. 22.83 ± 4.02ng/mL, p = 0.003). However, the mean umbilical cord serum leptin level in the pre-eclampsia group was significantly lower than in the control group (6.43 ± 2.08 vs. 7.27 ± 2.24; p = 0.034). There was a weak positive correlation between maternal serum leptin level and neonatal umbilical serum leptin level in the pre-eclamptic group (r = 0.21, p = 0.04). CONCLUSION Maternal serum leptin concentration is significantly increased in women with pre-eclampsia, compared with their normotensive counterparts. This increase becomes even more pronounced as the severity of the disease progresses. Maternal serum leptin assessment has the potential to become a veritable tool in the diagnosis and monitoring of pregnancies complicated by pre-eclampsia.
Collapse
Affiliation(s)
- Lazarus Ugochukwu Okafor
- Department of Obstetrics and Gynaecology, Nnamdi Azikiwe University Teaching Hospital (NAUTH), Nnewi, Nigeria
| | - George Uchenna Eleje
- Department of Obstetrics and Gynaecology, Nnamdi Azikiwe University Teaching Hospital (NAUTH), Nnewi, Nigeria
- Effective Care Research Unit, Department of Obstetrics and Gynaecology, Nnamdi Azikiwe University, Nnewi, Nigeria
| | - Joseph Ifeanyichukwu Ikechebelu
- Department of Obstetrics and Gynaecology, Nnamdi Azikiwe University Teaching Hospital (NAUTH), Nnewi, Nigeria
- Effective Care Research Unit, Department of Obstetrics and Gynaecology, Nnamdi Azikiwe University, Nnewi, Nigeria
| | - Emmanuel Onyebuchi Ugwu
- Department of Obstetrics and Gynaecology, College of Medicine, University of Nigeria Teaching Hospital, Enugu, Nigeria
| | - Obi Betrand Nwosu
- Department of Obstetrics and Gynaecology, Nnamdi Azikiwe University Teaching Hospital (NAUTH), Nnewi, Nigeria
| | - Okechukwu Christian Ikpeze
- Department of Obstetrics and Gynaecology, Nnamdi Azikiwe University Teaching Hospital (NAUTH), Nnewi, Nigeria
| | - Gerald Okanandu Udigwe
- Department of Obstetrics and Gynaecology, Nnamdi Azikiwe University Teaching Hospital (NAUTH), Nnewi, Nigeria
- Effective Care Research Unit, Department of Obstetrics and Gynaecology, Nnamdi Azikiwe University, Nnewi, Nigeria
| | - Ahizechukwu Chigoziem Eke
- Division of Maternal and Fetal Medicine, Department of Gynaecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | - Chukwudi Anthony Ogabido
- Department of Obstetrics and Gynaecology, Nnamdi Azikiwe University Teaching Hospital (NAUTH), Nnewi, Nigeria
| | | | | | - Chigozie Geoffrey Okafor
- Department of Obstetrics and Gynaecology, Nnamdi Azikiwe University Teaching Hospital (NAUTH), Nnewi, Nigeria
| |
Collapse
|
20
|
Marić I, Contrepois K, Moufarrej MN, Stelzer IA, Feyaerts D, Han X, Tang A, Stanley N, Wong RJ, Traber GM, Ellenberger M, Chang AL, Fallahzadeh R, Nassar H, Becker M, Xenochristou M, Espinosa C, De Francesco D, Ghaemi MS, Costello EK, Culos A, Ling XB, Sylvester KG, Darmstadt GL, Winn VD, Shaw GM, Relman DA, Quake SR, Angst MS, Snyder MP, Stevenson DK, Gaudilliere B, Aghaeepour N. Early prediction and longitudinal modeling of preeclampsia from multiomics. PATTERNS (NEW YORK, N.Y.) 2022; 3:100655. [PMID: 36569558 PMCID: PMC9768681 DOI: 10.1016/j.patter.2022.100655] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 09/28/2022] [Accepted: 11/11/2022] [Indexed: 12/13/2022]
Abstract
Preeclampsia is a complex disease of pregnancy whose physiopathology remains unclear. We developed machine-learning models for early prediction of preeclampsia (first 16 weeks of pregnancy) and over gestation by analyzing six omics datasets from a longitudinal cohort of pregnant women. For early pregnancy, a prediction model using nine urine metabolites had the highest accuracy and was validated on an independent cohort (area under the receiver-operating characteristic curve [AUC] = 0.88, 95% confidence interval [CI] [0.76, 0.99] cross-validated; AUC = 0.83, 95% CI [0.62,1] validated). Univariate analysis demonstrated statistical significance of identified metabolites. An integrated multiomics model further improved accuracy (AUC = 0.94). Several biological pathways were identified including tryptophan, caffeine, and arachidonic acid metabolisms. Integration with immune cytometry data suggested novel associations between immune and proteomic dynamics. While further validation in a larger population is necessary, these encouraging results can serve as a basis for a simple, early diagnostic test for preeclampsia.
Collapse
Affiliation(s)
- Ivana Marić
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mira N. Moufarrej
- Departments of Bioengineering and Applied Physics, Stanford University and Chan Zuckerberg Biohub, Stanford, CA 94305, USA
| | - Ina A. Stelzer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dorien Feyaerts
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xiaoyuan Han
- University of the Pacific, Arthur A. Dugoni School of Dentistry, San Francisco, CA 94103, USA
| | - Andy Tang
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Natalie Stanley
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ronald J. Wong
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gavin M. Traber
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mathew Ellenberger
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alan L. Chang
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ramin Fallahzadeh
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Huda Nassar
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Martin Becker
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Maria Xenochristou
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Camilo Espinosa
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Davide De Francesco
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mohammad S. Ghaemi
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Digital Technologies Research Centre, National Research Council Canada, Toronto, Canada
| | - Elizabeth K. Costello
- Departments of Medicine, and of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Anthony Culos
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xuefeng B. Ling
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Karl G. Sylvester
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gary L. Darmstadt
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Virginia D. Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gary M. Shaw
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David A. Relman
- Departments of Medicine, and of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Infectious Diseases Section, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Stephen R. Quake
- Departments of Bioengineering and Applied Physics, Stanford University and Chan Zuckerberg Biohub, Stanford, CA 94305, USA
| | - Martin S. Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David K. Stevenson
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brice Gaudilliere
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nima Aghaeepour
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
21
|
Abramova M, Churnosova M, Efremova O, Aristova I, Reshetnikov E, Polonikov A, Churnosov M, Ponomarenko I. Effects of Pre-Pregnancy Overweight/Obesity on the Pattern of Association of Hypertension Susceptibility Genes with Preeclampsia. Life (Basel) 2022; 12:2018. [PMID: 36556383 PMCID: PMC9784908 DOI: 10.3390/life12122018] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/21/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to explore the effects of pre-pregnancy overweight/obesity on the pattern of association of hypertension susceptibility genes with preeclampsia (PE). Ten single-nucleotide polymorphisms (SNPs) of the 10 genome-wide association studies (GWAS)-significant hypertension/blood pressure (BP) candidate genes were genotyped in 950 pregnant women divided into two cohorts according to their pre-pregnancy body mass index (preBMI): preBMI ≥ 25 (162 with PE and 159 control) and preBMI < 25 (290 with PE and 339 control). The PLINK software package was utilized to study the association (analyzed four genetic models using logistic regression). The functionality of PE-correlated loci was analyzed by performing an in silico database analysis. Two SNP hypertension/BP genes, rs805303 BAG6 (OR: 0.36−0.66) and rs167479 RGL3 (OR: 1.86), in subjects with preBMI ≥ 25 were associated with PE. No association between the studied SNPs and PE in the preBMI < 25 group was determined. Further analysis showed that two PE-associated SNPs are functional (have weighty eQTL, sQTL, regulatory, and missense values) and could be potentially implicated in PE development. In conclusion, this study was the first to discover the modifying influence of overweight/obesity on the pattern of association of GWAS-significant hypertension/BP susceptibility genes with PE: these genes are linked with PE in preBMI ≥ 25 pregnant women and are not PE-involved in the preBMI < 25 group.
Collapse
Affiliation(s)
- Maria Abramova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Maria Churnosova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Olesya Efremova
- Department of Medical Genetics, Kharkiv National Medical University, 61022 Kharkov, Ukraine
- Grishchenko Clinic of Reproductive Medicine, 61052 Kharkov, Ukraine
| | - Inna Aristova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Evgeny Reshetnikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Alexey Polonikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
- Department of Biology, Medical Genetics and Ecology and Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Irina Ponomarenko
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| |
Collapse
|
22
|
Fan M, Dong L, Meng Y, Wang Y, Zhen J, Qiu J. Leptin Promotes HTR-8/SVneo Cell Invasion via the Crosstalk between MTA1/WNT and PI3K/AKT Pathways. DISEASE MARKERS 2022; 2022:7052176. [PMID: 36457544 PMCID: PMC9708374 DOI: 10.1155/2022/7052176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 10/16/2022] [Accepted: 11/08/2022] [Indexed: 08/31/2023]
Abstract
The process of placental invasion is essential for a successful pregnancy. Leptin is involved in trophoblast invasiveness, and its dysregulation is connected with a series of diseases, including preeclampsia. However, the knowledge of the precise mechanisms in leptin-induced trophoblast invasiveness is still limited. According to the present research, transwell assay suggested that leptin is a dose- and time-dependent regulator in inducing HTR-8/SVneo cell invasion. Western blot analysis and immunofluorescence staining revealed that leptin-induced MMP9 expression is essential in the invasion process of HTR-8/SVneo cells. Mechanistically, we demonstrated that leptin activated β-catenin via the crosstalk between the MTA1/WNT and PI3K/AKT pathways. Besides, we showed that downregulating the key molecules in the signaling pathways by siRNA can inhibit leptin-induced MMP9 expression and further suppress invasion of HTR-8/SVneo cells. In conclusion, our study revealed a new regulatory mechanism of leptin-induced HTR-8/SVneo cell invasiveness and will provide novel insights into the causes and potential therapeutic targets for diseases related to dysregulation of trophoblast invasion in the future.
Collapse
Affiliation(s)
- Minghua Fan
- Department of Obstetrics and Gynecology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Lihua Dong
- Department of Obstetrics and Gynecology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yanping Meng
- Department of Obstetrics and Gynecology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Yao Wang
- Department of Obstetrics and Gynecology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Junhui Zhen
- Department of Pathology, School of Medicine, Shandong University, Jinan, 250021 Shandong, China
| | - Jianqing Qiu
- Department of Obstetrics and Gynecology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| |
Collapse
|
23
|
Grimaldi B, Kohan-Ghadr HR, Drewlo S. The Potential for Placental Activation of PPARγ to Improve the Angiogenic Profile in Preeclampsia. Cells 2022; 11:cells11213514. [PMID: 36359910 PMCID: PMC9659243 DOI: 10.3390/cells11213514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022] Open
Abstract
Preeclampsia (PE) is one of the most common causes of maternal-fetal morbidity and mortality world-wide. While the underlying causes of PE remain elusive, aberrant trophoblast differentiation and function are thought to cause an imbalance of secreted angiogenic proteins resulting in systemic endothelial dysfunction and organ damage in the mother. The placental dysfunction is also characterized by a reduction of the transcription factor, peroxisome proliferator activated receptor γ (PPARγ) which normally promotes trophoblast differentiation and healthy placental function. This study aimed to understand how placental activation of PPARγ effects the secretion of angiogenic proteins and subsequently endothelial function. To study this, healthy and PE placental tissues were cultured with or without the PPARγ agonist, Rosiglitazone, and a Luminex assay was performed to measure secreted proteins from the placenta. To assess the angiogenic effects of placental activation of PPARγ, human umbilical vein endothelial cells (HUVECs) were cultured with the placental conditioned media and the net angiogenic potential of these cells was measured by a tube formation assay. This is the first study to show PPARγ's beneficial effect on the angiogenic profile in the human preeclamptic placenta through the reduction of anti-angiogenic angiopoietin-2 and soluble endoglin and the upregulation of pro-angiogenic placental growth factor, fibroblast growth factor-2, heparin-binding epidermal growth factor, and follistatin. The changes in the angiogenic profile were supported by the increased angiogenic potential observed in the HUVECs when cultured with conditioned media from rosiglitazone-treated preeclamptic placentas. The restoration of these disrupted pathways by activation of PPARγ in the preeclamptic placenta offers potential to improve placental and endothelial function in PE.
Collapse
Affiliation(s)
- Brooke Grimaldi
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Hamid-Reza Kohan-Ghadr
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Sascha Drewlo
- Biological Sciences Platform, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto M4N 3M5, Canada
- Department of Obstetrics and Gynecology, Temerty Faculty of Medicine, University of Toronto, Toronto M5G 1E2, Canada
- Correspondence:
| |
Collapse
|
24
|
Danielli M, Thomas RC, Gillies CL, Hu J, Khunti K, Tan BK. Blood biomarkers to predict the onset of pre-eclampsia: A systematic review and meta-analysis. Heliyon 2022; 8:e11226. [PMID: 36387521 PMCID: PMC9649987 DOI: 10.1016/j.heliyon.2022.e11226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/29/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Pre-eclampsia is one of the most common pregnancy complications, and a major cause of fetal and maternal morbidity and mortality globally. Diagnosis currently takes place in the third trimester based on clinical symptoms. This systematic review and meta-analysis sought to determine the blood biomarkers that are associated with pre-eclampsia, and in particular, the biomarkers that could predict pre-eclampsia in early pregnancy. We searched the electronic databases (Medline, Embase, Cochrane Library) from inception up to March 2022. Prospective studies with 1000 or more participants that measured blood biomarkers to predict or diagnose pre-eclampsia have been included in this systematic review. Biomarkers' measurements were considered from the first up to the third trimester, but not during labor. Data concerning pre-eclampsia, biomarker measurements and study characteristics were extracted. Meta-analysis was performed when possible. We found a total of 43 studies (assessing 62 different biomarkers in 18,170 pregnancies, have been included in this systematic review, and a total of 6 studies (assessing 2 biomarkers have been included in the meta-analysis). Statistical analysis was performed for PlGF and sFlt-1. Mean difference in PlGF levels between pre-eclampsia and healthy pregnancies, appear to increase as the pregnancy progresses. Results of sFlt-1 meta-analysis were inconclusive. No significant publication bias was identified. This is the most comprehensive and up to date systematic review and meta-analysis on this important topic on blood biomarkers for the early prediction of pre-eclampsia. Further This research highlights the urgent needed for further discovery research to identify blood biomarkers that could predict the development of pre-eclampsia.
Collapse
Affiliation(s)
- Marianna Danielli
- Cardiovascular Sciences, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Roisin C. Thomas
- Cardiovascular Sciences, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Clare L. Gillies
- Diabetes Research Centre, Leicester General Hospital, Leicester, LE5 4PW, United Kingdom
| | - Jiamiao Hu
- Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, Fujian, China
| | - Kamlesh Khunti
- Diabetes Research Centre, Leicester General Hospital, Leicester, LE5 4PW, United Kingdom
- NIHR Applied Research Collaboration – East Midlands (ARC-EM), Leicester General Hospital, Leicester, LE5 4PW, United Kingdom
| | - Bee Kang Tan
- Cardiovascular Sciences, University of Leicester, Leicester, LE1 7RH, United Kingdom
- Diabetes Research Centre, Leicester General Hospital, Leicester, LE5 4PW, United Kingdom
| |
Collapse
|
25
|
Chen X, Liu Z, Cui J, Chen X, Xiong J, Zhou W. Circulating adipokine levels and preeclampsia: A bidirectional Mendelian randomization study. Front Genet 2022; 13:935757. [PMID: 36072663 PMCID: PMC9444139 DOI: 10.3389/fgene.2022.935757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/22/2022] [Indexed: 12/02/2022] Open
Abstract
Background: Several observational studies have demonstrated that significantly rising circulating adipokine levels are pervasive in preeclampsia or eclampsia disorder (or preeclampsia toxemia (PET)). However, it remains unclear whether this relationship is causal. In this study, we sought to elucidate the causal effects of circulating adipokine levels on PET. Methods: Summary-level data and independent genetic variants strongly associated with common adipokine molecule (adiponectin, leptin, resistin, sOB-R, and PAI-1) levels were drawn from public genome-wide association study (GWASs). Additionally, the corresponding effects between instrumental variables and PET outcomes were acquired from the FinnGen consortium, including 4,743 cases and 136,325 controls of European ancestry. Subsequently, an inverse-variance weighted (IVW) approach was applied for the principal two-sample Mendelian randomization (MR) and multivariable MR (MVMR) analyses. Various complementary sensitivity analyses were then carried out to determine the robustness of our models. Results: The results of the IVW method did not reveal any causal relationship shared across genetically predisposed adipokine levels and PET risk (for adiponectin, OR = 0.86, 95% CI: 0.65–1.13, p = 0.274). Additionally, no significant associations were identified after taking into account five circulating adipokines in MVMR research. Complementary sensitivity analysis also supported no significant associations between them. In the reverse MR analysis, genetically predicted PET risk showed a suggestive association with elevating PAI-1 levels by the IVW method (Beta = 0.120, 95% CI: 0.014, 0.227, p = 0.026). Furthermore, there were no strong correlations between genetic liability to PET and other adipokine levels (p > 0.05). Conclusion: Our MR study did not provide robust evidence supporting the causal role of common circulating adipokine levels in PET, whereas genetically predicted PET may instrumentally affect PAI-1 levels. These findings suggest that PAI-1 may be a useful biomarker for monitoring the diagnosis or therapy of PET rather than a therapeutic target for PET.
Collapse
|
26
|
Faulkner JL, Wright D, Antonova G, Jaffe IZ, Kennard S, Belin de Chantemèle EJ. Midgestation Leptin Infusion Induces Characteristics of Clinical Preeclampsia in Mice, Which Is Ablated by Endothelial Mineralocorticoid Receptor Deletion. Hypertension 2022; 79:1536-1547. [PMID: 35510543 DOI: 10.1161/hypertensionaha.121.18832] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Patients with preeclampsia demonstrate increases in placental leptin production in midgestation, and an associated increase in late gestation plasma leptin levels. The consequences of mid-late gestation increases in leptin production in pregnancy is unknown. Our previous work indicates that leptin infusion induces endothelial dysfunction in nonpregnant female mice via leptin-mediated aldosterone production and endothelial mineralocorticoid receptor (ECMR) activation, which is ablated by ECMR deletion. Therefore, we hypothesized that leptin infusion in mid-gestation of pregnancy induces endothelial dysfunction and hypertension, hallmarks of clinical preeclampsia, which are prevented by ECMR deletion. METHODS Leptin was infused via miniosmotic pump (0.9 mg/kg per day) into timed-pregnant ECMR-intact (WT) and littermate-mice with ECMR deletion (KO) on gestation day (GD)11-18. RESULTS Leptin infusion decreased fetal weight and placental efficiency in WT mice compared with WT+vehicle. Radiotelemetry recording demonstrated that blood pressure increased in leptin-infused WT mice during infusion. Leptin infusion reduced endothelial-dependent relaxation responses to acetylcholine (ACh) in both resistance (second-order mesenteric) and conduit (aorta) vessels in WT pregnant mice. Leptin infusion increased placental ET-1 (endothelin-1) production evidenced by increased PPET-1 (preproendothelin-1) and ECE-1 (endothelin-converting enzyme-1) expressions in WT mice. Adrenal aldosterone synthase (CYP11B2) and angiotensin II type 1 receptor b (AT1Rb) expression increased with leptin infusion in pregnant WT mice. KO pregnant mice demonstrated protection from leptin-induced reductions in pup weight, placental efficiency, increased BP, and endothelial dysfunction. CONCLUSIONS Collectively, these data indicate that leptin infusion in midgestation induces endothelial dysfunction, hypertension, and fetal growth restriction in pregnant mice, which is ablated by ECMR deletion.
Collapse
Affiliation(s)
- Jessica L Faulkner
- Department of Physiology (J.L.F.), Medical College of Georgia at Augusta University
| | - Derrian Wright
- Vascular Biology Center (D.W., G.A., S.K., E.J.B.d.C.), Medical College of Georgia at Augusta University
| | - Galina Antonova
- Vascular Biology Center (D.W., G.A., S.K., E.J.B.d.C.), Medical College of Georgia at Augusta University
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (I.Z.J.)
| | - Simone Kennard
- Vascular Biology Center (D.W., G.A., S.K., E.J.B.d.C.), Medical College of Georgia at Augusta University
| | - Eric J Belin de Chantemèle
- Vascular Biology Center (D.W., G.A., S.K., E.J.B.d.C.), Medical College of Georgia at Augusta University.,Department of Cardiology (E.J.B.d.C.), Medical College of Georgia at Augusta University
| |
Collapse
|
27
|
Gyselaers W, Lees C. Maternal Low Volume Circulation Relates to Normotensive and Preeclamptic Fetal Growth Restriction. Front Med (Lausanne) 2022; 9:902634. [PMID: 35755049 PMCID: PMC9218216 DOI: 10.3389/fmed.2022.902634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
This narrative review summarizes current evidence on the association between maternal low volume circulation and poor fetal growth. Though much work has been devoted to the study of cardiac output and peripheral vascular resistance, a low intravascular volume may explain why high vascular resistance causes hypertension in women with preeclampsia (PE) that is associated with fetal growth restriction (FGR) and, at the same time, presents with normotension in FGR itself. Normotensive women with small for gestational age babies show normal gestational blood volume expansion superimposed upon a constitutionally low intravascular volume. Early onset preeclampsia (EPE; occurring before 32 weeks) is commonly associated with FGR, and poor plasma volume expandability may already be present before conception, thus preceding gestational volume expansion. Experimentally induced low plasma volume in rodents predisposes to poor fetal growth and interventions that enhance plasma volume expansion in FGR have shown beneficial effects on intrauterine fetal condition, prolongation of gestation and birth weight. This review makes the case for elevating the maternal intravascular volume with physical exercise with or without Nitric Oxide Donors in FGR and EPE, and evaluating its role as a potential target for prevention and/or management of these conditions.
Collapse
Affiliation(s)
- Wilfried Gyselaers
- Department of Obstetrics, Ziekenhuis Oost Limburg, Genk, Belgium.,Department of Physiology, Hasselt University, Hasselt, Belgium
| | - Christoph Lees
- Centre for Fetal Care, Queen Charlotte's and Chelsea Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom.,Department of Metabolism, Digestion and Reproduction, Institute for Reproductive and Developmental Biology, Imperial College London, London, United Kingdom.,Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Centre for Fetal Care, Queen Charlotte's and Chelsea Hospital, London, United Kingdom
| |
Collapse
|
28
|
Karpuzoglu H, Ucal Y, Kumru P, Muhcu M, Eroglu M, Serdar M, Serteser M, Ozpinar A. Increased maternal leptin levels may be an indicator of subclinical hypothyroidism in a newborn. J Med Biochem 2022; 41:156-161. [PMID: 35510203 PMCID: PMC9010041 DOI: 10.5937/jomb0-32425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 09/01/2021] [Indexed: 12/03/2022] Open
Abstract
Background Several factors may influence newborn thyroid-stimulating hormone (TSH) concentrations and cause subclinical hypothyroidism in a newborn. A sufficient level of leptin signalling is needed for the normal production of TSH and thyroid hormones by the thyroid gland. Our study aimed to investigate the correlation between maternal serum leptin concentration during the third trimester of pregnancy and newborn screening-TSH levels. Methods This prospective cross-sectional study was conducted in obstetrics and gynaecology clinics of a state hospital between June and August 2013. Maternal venous blood samples were collected from 270 healthy pregnant women in the third trimester just before delivery. Measurements of maternal fT3, fT4, TSH, anti-thyroid peroxidase (TPO), and anti-thyroglobulin (anti-Tg) antibodies from serum samples were performed by chemiluminescence immunoassay. Maternal serum leptin levels were determined by ELISA. Dried capillary blood spots were used to measure newborn TSH levels. Results Subjects were divided into two groups according to the neonatal TSH levels using a cut-point of 5.5 mIU/L. Median maternal serum leptin levels were significantly higher in newborns whose TSH levels were higher than >5.5 mIU/L [13.2 μg/L (1.3 - 46.5) vs 19.7 μg/L (2.4 - 48.5), p<0.05]. Serum leptin levels showed a negative correlation with maternal fT4 (r=0.32, p<0.05), fT3 (r=0.23, p<0.05), and a positive correlation with BMI (r=0.30, p<0.05). Conclusions Our results suggest that high leptin levels in the third trimester of pregnancy influence maternal thyroid functions and might cause an increase in newborn TSH levels. Detection of high maternal serum leptin levels may be a reason for subclinical hypothyroidism.
Collapse
Affiliation(s)
- Hande Karpuzoglu
- Acibadem Mehmet Ali Aydinlar University, School of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
| | - Yasemin Ucal
- Acibadem Mehmet Ali Aydinlar University, School of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
| | - Pinar Kumru
- Zeynep Kamil Research and Training Hospital, Department of Obstetrics and Gynecology, Istanbul, Turkey
| | - Murat Muhcu
- Umraniye Training and Research Hospital, Department of Obstetrics and Gynecology, Istanbul, Turkey
| | - Mustafa Eroglu
- Haydarpasa Numune Training and Research Hospital, Department of Obstetrics and Gynecology, Istanbul, Turkey
| | - Muhittin Serdar
- Acibadem Mehmet Ali Aydinlar University, School of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
| | - Mustafa Serteser
- Acibadem Mehmet Ali Aydinlar University, School of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
| | - Aysel Ozpinar
- Acibadem Mehmet Ali Aydinlar University, School of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
| |
Collapse
|
29
|
Furigo IC, Dearden L. Mechanisms mediating the impact of maternal obesity on offspring hypothalamic development and later function. Front Endocrinol (Lausanne) 2022; 13:1078955. [PMID: 36619540 PMCID: PMC9813846 DOI: 10.3389/fendo.2022.1078955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
As obesity rates have risen around the world, so to have pregnancies complicated by maternal obesity. Obesity during pregnancy is not only associated with negative health outcomes for the mother and the baby during pregnancy and birth, there is also strong evidence that exposure to maternal obesity causes an increased risk to develop obesity, diabetes and cardiovascular disease later in life. Animal models have demonstrated that increased weight gain in offspring exposed to maternal obesity is usually preceded by increased food intake, implicating altered neuronal control of food intake as a likely area of change. The hypothalamus is the primary site in the brain for maintaining energy homeostasis, which it coordinates by sensing whole body nutrient status and appropriately adjusting parameters including food intake. The development of the hypothalamus is plastic and regulated by metabolic hormones such as leptin, ghrelin and insulin, making it vulnerable to disruption in an obese in utero environment. This review will summarise how the hypothalamus develops, how maternal obesity impacts on structure and function of the hypothalamus in the offspring, and the factors that are altered in an obese in utero environment that may mediate the permanent changes to hypothalamic function in exposed individuals.
Collapse
Affiliation(s)
- Isadora C. Furigo
- Centre for Sport, Exercise and Life Sciences, School of Life Sciences, Coventry University, Coventry, United Kingdom
| | - Laura Dearden
- Metabolic Research Laboratories, Wellcome MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Laura Dearden,
| |
Collapse
|
30
|
Olerich K, Soper D, Delaney S, Sterrett M. Pregnancy Care for Patients With Super Morbid Obesity. Front Pediatr 2022; 10:839377. [PMID: 35928678 PMCID: PMC9343711 DOI: 10.3389/fped.2022.839377] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
The patient with obesity represents unique challenges to the medical community and, in the setting of pregnancy, additional risks to both mother and fetus. This document will focus on the risks and considerations needed to care for the women with obesity and her fetus during the antepartum, intrapartum, and immediate postpartum stages of pregnancy. Specific attention will be given to pregnancy in the setting of class III and super morbid obesity.
Collapse
Affiliation(s)
- Kelsey Olerich
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - David Soper
- Department of Obstetrics and Gynecology, Medical University of South Carolina, Charleston, SC, United States
| | - Shani Delaney
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Mary Sterrett
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States.,Department of Obstetrics and Gynecology, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
31
|
Y Al-Taee SM, Al-Allaff RGM, E Alnajafy L. The Effect of Leptin on the Regulation of Immune Responses in Women with Polycystic Ovary Syndrome. Pak J Biol Sci 2022; 25:715-724. [PMID: 36098197 DOI: 10.3923/pjbs.2022.715.724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
<b>Background and Objective:</b> Leptin is a hormone produced by fat cells in adipose tissue that plays a biological role in metabolism, immune system control and energy flow regulation. The study's objective was to investigate serum leptin levels in PCOS females and determine how they are related to immunological and hormonal parameters. <b>Materials and Methods:</b> Thirty PCOS women were chosen for the study and compared to thirty healthy women as control samples, with both case and normal samples ranging in age from 18-36 years. WBC count, absolute numbers of neutrophils, lymphocytes, monocytes, eosinophils, leptin, Prolactin and serum IgA levels were measured. <b>Results:</b> The results showed the mean WBC, lymphocytes and eosinophil absolute numbers in PCOS women were significantly different. However, there was no significant difference in the absolute numbers of neutrophils and monocytes. The findings also revealed a significant increase in BMI, IgA, leptin and prolactin levels in PCOS when compared to controls. The results showed strong positive correlation coefficients between BMI and IgA (1.000**), WBC with neutrophils, lymphocytes and monocytes (0.797**, 0.790**, 0.712**), respectively and finally leptin and prolactin (0.474**). The same test, on the other hand, revealed an inverse correlation coefficient between BMI, IgA and prolactin (-0.376*, -0.376*, respectively with p<u><</u>0.05). <b>Conclusion:</b> A rise in the levels of the hormones leptin and prolactin, which were positively associated with the body mass index in women with PCOS, was found in the current study. The findings revealed that the hormones leptin and prolactin have an effect on some immune parameters in women with PCOS.
Collapse
|
32
|
Obeidat RA, Abdo N, Sakee B, Alghazo S, Jbarah OF, Hazaimeh EA, Albeitawi S. Maternal and fetal serum leptin levels and their association with maternal and fetal variables and labor: A cross-sectional study. Ann Med Surg (Lond) 2021; 72:103050. [PMID: 34815864 PMCID: PMC8591461 DOI: 10.1016/j.amsu.2021.103050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/06/2021] [Accepted: 11/09/2021] [Indexed: 12/24/2022] Open
Abstract
Background Leptin is a polypeptide hormone that may be implicated in the pathogenesis of various disorders during pregnancy. We sought to determine serum leptin levels among pregnant women and their fetuses and to investigate their association with fetal and maternal variables. Method 452 pregnant women who attended to labor ward between January 2020 and August 2020 were included in the study. Serum leptin concentrations were measured using enzyme-linked immunosorbent assay method. Mann-Whitney U test and Spearman's correlation test were used for statistical analysis. A multivariate linear regression analysis was then performed. Significance level was considered at alpha <0.05. Results The median maternal and fetal serum leptin levels were 6.42 [4.16-8.51] ng/mL and 2.9 [1.03-5.36] ng/mL respectively. There was no significant correlation between maternal and fetal serum leptin levels (p = 0.064). Maternal serum leptin levels correlated positively with maternal body mass index (BMI) (r = 0.117, p = 0.005). Besides, maternal serum leptin levels were significantly higher in nulliparous women (7.57 [4.45-9.30] ng/mL vs. 6.22 [4.02-8.30] ng/mL, p = 0.037) and in women who were in active labor (6.83 [4.39-8.92] ng/mL vs. 6.25 [4.04-8.30] ng/mL, p = 0.047). Fetal serum leptin levels were significantly higher in large for gestational age (LGA) fetuses (4.81 [2.13-7.22] ng/mL vs. 2.80 [0.96-5.16] ng/mL, p = 0.003) and in fetuses with preterm premature ruptures of membranes (PPROM) (5.23 [2.42-8.07] ng/mL vs. 2.86 [1.00-5.23] ng/mL, p = 0.021). Conclusion Maternal serum leptin levels were influenced by maternal BMI, parity and labor. Fetal serum leptin levels were higher among LGA fetuses and in fetuses with PPROM.
Collapse
Affiliation(s)
- Rawan A Obeidat
- Department of Obstetrics and Gynecology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Nour Abdo
- Department of Public Health and Family Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Baraa Sakee
- Department of Obstetrics and Gynecology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Shahed Alghazo
- Department of Obstetrics and Gynecology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Omar F Jbarah
- Department of Neuroscience, Neurosurgery Division, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Ethar A Hazaimeh
- Department of Neuroscience, Neurology Division, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Soha Albeitawi
- Department of Obstetrics and Gynecology, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| |
Collapse
|
33
|
Huang Q, Hao S, You J, Yao X, Li Z, Schilling J, Thyparambil S, Liao WL, Zhou X, Mo L, Ladella S, Davies-Balch SR, Zhao H, Fan D, Whitin JC, Cohen HJ, McElhinney DB, Wong RJ, Shaw GM, Stevenson DK, Sylvester KG, Ling XB. Early-pregnancy prediction of risk for pre-eclampsia using maternal blood leptin/ceramide ratio: discovery and confirmation. BMJ Open 2021; 11:e050963. [PMID: 34824115 PMCID: PMC8627403 DOI: 10.1136/bmjopen-2021-050963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE This study aimed to develop a blood test for the prediction of pre-eclampsia (PE) early in gestation. We hypothesised that the longitudinal measurements of circulating adipokines and sphingolipids in maternal serum over the course of pregnancy could identify novel prognostic biomarkers that are predictive of impending event of PE early in gestation. STUDY DESIGN Retrospective discovery and longitudinal confirmation. SETTING Maternity units from two US hospitals. PARTICIPANTS Six previously published studies of placental tissue (78 PE and 95 non-PE) were compiled for genomic discovery, maternal sera from 15 women (7 non-PE and 8 PE) enrolled at ProMedDx were used for sphingolipidomic discovery, and maternal sera from 40 women (20 non-PE and 20 PE) enrolled at Stanford University were used for longitudinal observation. OUTCOME MEASURES Biomarker candidates from discovery were longitudinally confirmed and compared in parallel to the ratio of placental growth factor (PlGF) and soluble fms-like tyrosine kinase (sFlt-1) using the same cohort. The datasets were generated by enzyme-linked immunosorbent and liquid chromatography-tandem mass spectrometric assays. RESULTS Our discovery integrating genomic and sphingolipidomic analysis identified leptin (Lep) and ceramide (Cer) (d18:1/25:0) as novel biomarkers for early gestational assessment of PE. Our longitudinal observation revealed a marked elevation of Lep/Cer (d18:1/25:0) ratio in maternal serum at a median of 23 weeks' gestation among women with impending PE as compared with women with uncomplicated pregnancy. The Lep/Cer (d18:1/25:0) ratio significantly outperformed the established sFlt-1/PlGF ratio in predicting impending event of PE with superior sensitivity (85% vs 20%) and area under curve (0.92 vs 0.52) from 5 to 25 weeks of gestation. CONCLUSIONS Our study demonstrated the longitudinal measurement of maternal Lep/Cer (d18:1/25:0) ratio allows the non-invasive assessment of PE to identify pregnancy at high risk in early gestation, outperforming the established sFlt-1/PlGF ratio test.
Collapse
Affiliation(s)
| | - Shiying Hao
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California, USA
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Palo Alto, California, USA
| | - Jin You
- Department of Bioengineering, University of California Riverside, Riverside, California, USA
| | | | - Zhen Li
- Department of Surgery, Stanford University, Stanford, California, USA
- Binhai Industrial Technology Research Institute, Zhejiang University, Tianjin, China
- School of Electrical Engineering, Southeast University, Nanjing, China
| | | | | | | | - Xin Zhou
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China
| | - Lihong Mo
- Department of Obstetrics and Gynecology, University of California San Francisco, Fresno, California, USA
| | - Subhashini Ladella
- Department of Obstetrics and Gynecology, University of California San Francisco, Fresno, California, USA
| | | | - Hangyi Zhao
- Department of Mathematics, Stanford University, Stanford, California, USA
| | - David Fan
- Department of Statistics and Applied Probability, University of California Santa Barbara, Santa Barbara, California, USA
| | - John C Whitin
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Harvey J Cohen
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Doff B McElhinney
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California, USA
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Palo Alto, California, USA
| | - Ronald J Wong
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Gary M Shaw
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - David K Stevenson
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Karl G Sylvester
- Department of Surgery, Stanford University, Stanford, California, USA
| | - Xuefeng B Ling
- Department of Surgery, Stanford University, Stanford, California, USA
| |
Collapse
|
34
|
Miao T, Yu Y, Sun J, Ma A, Yu J, Cui M, Yang L, Wang H. Decrease in abundance of bacteria of the genus Bifidobacterium in gut microbiota may be related to pre-eclampsia progression in women from East China. Food Nutr Res 2021; 65:5781. [PMID: 34262418 PMCID: PMC8254465 DOI: 10.29219/fnr.v65.5781] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/15/2021] [Accepted: 02/02/2021] [Indexed: 01/08/2023] Open
Abstract
Background Pre-eclampsia (PE) can result in severe damage to maternal and fetal health. It has been reported that gut microbiota (GM) had important roles in regulating the metabolic and inflammatory responses of the mother. However, investigations on GM in PE are rare. Objective The objective of the present study was to investigate the changes of GM in PE and how to alter the GM composition in PE by dietary or dietary supplements. Design We analyzed the composition changes in GM as well as the relationship between bacteria of different genera and clinical indices by amplifying the V4 region of the 16S ribosomal RNA gene in 12 PE patients and eight healthy pregnant women in East China. Results In the PE group, the Observed Species Index was lower than that in the control group, indicating that the α-diversity of the microbiome in the PE group decreased. At phylum, family, and genus levels, the relative abundance of different bacteria in PE patients displayed substantial differences to those from healthy women. We noted a decreased abundance of bacteria of the phylum Actinobacteria (P = 0.042), decreased abundance of bacteria of the family Bifidobacteriaceae (P = 0.039), increased abundance of bacteria of the genus Blautia (P = 0.026) and Ruminococcus (P = 0.048), and decreased abundance of bacteria of the genus Bifidobacterium (P = 0.038). Among three enriched genera, bacteria of the genus Bifidobacterium showed a negative correlation with the systolic blood pressure (SBP), diastolic blood pressure (DBP), and dyslipidemia, which involved glucose metabolism, lipid metabolism, and the oxidative-phosphorylation pathway. The increased abundance of bacteria of the genera Blautia and Ruminococcus was positively correlated with obesity and dyslipidemia, which involved lipid metabolism, glycosyltransferases, biotin metabolism, and the oxidative-phosphorylation pathways. Moreover, women in the PE group ate more than women in the control group, so fetuses were more prone to overnutrition in the PE group. Conclusion There is a potential for GM dysbiosis in PE patients, and they could be prone to suffer from metabolic syndrome. We speculate that alterations in the abundance of bacteria of certain genera (e.g. increased abundance of Blautia and Ruminococcus, and decreased abundance of Bifidobacterium) were associated with PE development to some degree. Our data could help to monitor the health of pregnant women and may be helpful for preventing and assisting treatment of PE by increasing dietary fiber or probiotics supplement.
Collapse
Affiliation(s)
- Tingting Miao
- Department of Education, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, China.,Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Yun Yu
- Department of Clinical Laboratory, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Aiguo Ma
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Jinran Yu
- School of Public Health, Qingdao University, Qingdao, China
| | - Mengjun Cui
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Liping Yang
- Department of Obstetrics and Gynecology, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Huiyan Wang
- Department of Obstetrics and Gynecology, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, China
| |
Collapse
|
35
|
Serebrova VN, Trifonova EA, Stepanov VA. Natural Selection as a Driver for the Genetic Component of Preeclampsia. Mol Biol 2021. [DOI: 10.1134/s0026893321020308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
36
|
de Knegt VE, Hedley PL, Kanters JK, Thagaard IN, Krebs L, Christiansen M, Lausten-Thomsen U. The Role of Leptin in Fetal Growth during Pre-Eclampsia. Int J Mol Sci 2021; 22:ijms22094569. [PMID: 33925454 PMCID: PMC8123779 DOI: 10.3390/ijms22094569] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
Leptin is secreted by the placenta and has a multi-facetted role in the regulation of functions related to pregnancy. Metabolic disorders and insufficient homeostatic compensatory mechanisms involving leptin during pregnancy play a decisive role in the development of pre-eclampsia (PE) and give rise to compromised intrauterine growth conditions and aberrant birth weight of offspring. This review was compiled to elucidate the metabolic background of PE and its relationship with adverse intrauterine growth conditions through the examination of leptin as well as to describe possible mechanisms linking leptin to fetal growth restriction. This review illustrates that leptin in PE is dysregulated in maternal, fetal, and placental compartments. There is no single set of unifying mechanisms within the spectrum of PE, and regulatory mechanisms involving leptin are specific to each situation. We conclude that dysregulated leptin is involved in fetal growth at many levels through complex interactions with parallel pregnancy systems and probably throughout the entirety of pregnancy.
Collapse
Affiliation(s)
- Victoria E. de Knegt
- Department for Congenital Disorders, Danish National Biobank and Biomarkers, Statens Serum Institute, Artillerivej 5, 2300 Copenhagen, Denmark; (P.L.H.); (M.C.)
- Correspondence: ; Tel.: +45-50469429
| | - Paula L. Hedley
- Department for Congenital Disorders, Danish National Biobank and Biomarkers, Statens Serum Institute, Artillerivej 5, 2300 Copenhagen, Denmark; (P.L.H.); (M.C.)
| | - Jørgen K. Kanters
- Laboratory of Experimental Cardiology, Department of Biomedical Science, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark;
| | - Ida N. Thagaard
- Department of Gynecology and Obstetrics, Copenhagen University Hospital Slagelse, Ingemannsvej 18, 4200 Slagelse, Denmark;
| | - Lone Krebs
- Department of Obstetrics and Gynecology, University Hospital Hvidovre, Kettegård Alle 30, 2650 Hvidovre, Denmark;
| | - Michael Christiansen
- Department for Congenital Disorders, Danish National Biobank and Biomarkers, Statens Serum Institute, Artillerivej 5, 2300 Copenhagen, Denmark; (P.L.H.); (M.C.)
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Ulrik Lausten-Thomsen
- Department of Neonatology, University Hospital Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark;
| |
Collapse
|
37
|
Xu H, Xie Y, Sun Y, Guo R, Lv D, Li X, Li F, He M, Fan Y, Deng D. Integrated analysis of multiple microarray studies to identify potential pathogenic gene modules in preeclampsia. Exp Mol Pathol 2021; 120:104631. [PMID: 33744280 DOI: 10.1016/j.yexmp.2021.104631] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 01/15/2021] [Accepted: 03/14/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND Preeclampsia is a life-threatening hypertensive disorder during pregnancy, while underlying pathogenesis and its diagnosis are incomplete. METHODS In this study, we utilized the Robust Rank Aggregation method to integrate 6 eligible preeclampsia microarray datasets from Gene Expression Omnibus database. We used linear regression to assess the associations between significant differentially expressed genes (DEGs) and blood pressure. Functional annotation, protein-protein interaction, Gene Set Enrichment Analysis (GSEA) and single sample GSEA were employed for investigating underlying pathogenesis in preeclampsia. RESULTS We filtered 52 DEGs and further screened for 5 hub genes (leptin, pappalysin 2, endoglin, fms related receptor tyrosine kinase 1, tripartite motif containing 24) that were positively correlated with both systolic blood pressure and diastolic blood pressure. Receiver operating characteristic indicated that hub genes were potential biomarkers for diagnosis and prognosis in preeclampsia. GSEA for single hub gene revealed that they were all closely related to angiogenesis and estrogen response in preeclampsia. Moreover, single sample GSEA showed that the expression levels of 5 hub genes were correlated with those of immune cells in immunologic microenvironment at maternal-fetal interface. CONCLUSIONS These findings provide new insights into underlying pathogenesis in preeclampsia; 5 hub genes were identified as biomarkers for diagnosis and prognosis in preeclampsia.
Collapse
Affiliation(s)
- Heze Xu
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; The Second Clinical Medicine College, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, China
| | - Yin Xie
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yanan Sun
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Rong Guo
- Department of Software Engineering College, Information and Computer Engineering, Northeast Forestry University, Harbin, Heilongjiang, China
| | - Dan Lv
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuanxuan Li
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fanfan Li
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mengzhou He
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yao Fan
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dongrui Deng
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
38
|
Palei AC, Martin HL, Wilson BA, Anderson CD, Granger JP, Spradley FT. Impact of hyperleptinemia during placental ischemia-induced hypertension in pregnant rats. Am J Physiol Heart Circ Physiol 2021; 320:H1949-H1958. [PMID: 33710923 DOI: 10.1152/ajpheart.00724.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The prevalence of preeclampsia and obesity have increased. Although obesity is a major risk factor for preeclampsia, the mechanisms linking these morbidities are poorly understood. Circulating leptin levels increase in proportion to fat mass. Infusion of this adipokine elicits hypertension in nonpregnant rats, but less is known about how hyperleptinemia impacts blood pressure during placental ischemia, an initiating event in the pathophysiology of hypertension in preeclampsia. We tested the hypothesis that hyperleptinemia during reduced uterine perfusion pressure (RUPP) exaggerates placental ischemia-induced hypertension. On gestational day (GD) 14, Sprague-Dawley rats were implanted with osmotic mini-pumps delivering recombinant rat leptin (1 µg/kg/min iv) or vehicle concurrently with the RUPP procedure to induce placental ischemia or Sham. On GD 19, plasma leptin was elevated in Sham + Leptin and RUPP + Leptin. Leptin infusion did not significantly impact mean arterial pressure (MAP) in Sham. MAP was increased in RUPP + Vehicle vs. Sham + Vehicle. In contrast to our hypothesis, placental ischemia-induced hypertension was attenuated by leptin infusion. To examine potential mechanisms for attenuation of RUPP-induced hypertension during hyperleptinemia, endothelial-dependent vasorelaxation to acetylcholine was similar between Sham and RUPP; however, endothelial-independent vasorelaxation to the nitric oxide (NO)-donor, sodium nitroprusside, was increased in Sham and RUPP. These findings suggest that NO/cyclic guanosine monophosphate (cGMP) signaling was increased in the presence of hyperleptinemia. Plasma cGMP was elevated in Sham and RUPP hyperleptinemic groups compared with vehicle groups but plasma and vascular NO metabolites were reduced. These data suggest that hyperleptinemia during placental ischemia attenuates hypertension by compensatory increases in NO/cGMP signaling.NEW & NOTEWORTHY Ours is the first study to examine the impact of hyperleptinemia on the development of placental ischemia-induced hypertension using an experimental animal model.
Collapse
Affiliation(s)
- Ana C Palei
- Department of Surgery, The University of Mississippi Medical Center, Jackson, Mississippi
| | - Hunter L Martin
- Department of Surgery, The University of Mississippi Medical Center, Jackson, Mississippi.,Department of Physiology and Biophysics, The University of Mississippi Medical Center, Jackson, Mississippi
| | - Barbara A Wilson
- Department of Surgery, The University of Mississippi Medical Center, Jackson, Mississippi
| | - Christopher D Anderson
- Department of Surgery, The University of Mississippi Medical Center, Jackson, Mississippi
| | - Joey P Granger
- Department of Physiology and Biophysics, The University of Mississippi Medical Center, Jackson, Mississippi
| | - Frank T Spradley
- Department of Surgery, The University of Mississippi Medical Center, Jackson, Mississippi.,Department of Physiology and Biophysics, The University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
39
|
Kapustin RV, Kopteeva EV, Alekseenkova EN, Tral TG, Tolibova GK, Arzhanova ON. Placental expression of endoglin, placental growth factor, leptin, and hypoxia-inducible factor-1 in diabetic pregnancy and pre-eclampsia. Gynecol Endocrinol 2021; 37:35-39. [PMID: 34937509 DOI: 10.1080/09513590.2021.2006513] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
OBJECTIVE To evaluate a level of expression of endoglin (Eng), leptin (Lep), placental growth factor (PlGF), and hypoxia-inducible factor-1alpha (HIF-1α) in placenta among women with pre-eclampsia and diabetes mellitus (DM), considering the method of glycemia correction and preconception care. MATERIALS AND METHODS A retrospective cohort study was conducted. A total of 124 women were divided into following groups: type 1 DM (n = 40), type 2DM (n = 31), gestational DM (n = 33), pre-eclampsia without DM (PE) (n = 10) and the control group (n = 10). The histochemical study was performed by using primary monoclonal antibodies to Eng, PlGF, Lep, and HIF-1α (Abcam, UK). RESULTS The highest level of placental expression of Eng was observed in the PE group (20.34%). The same trend was also typical for T1DM (not planned) and insulin-treated groups: T2DM and GDM. An amount of cell with an PlGF expression was significantly higher in the control group (12.2%), while the lowest was observed in the pre-eclampsia group (1.18%) and T1DM (not planned) (1.26%). The placental leptin expression within each DM group was increased among the patients with unplanned pregnancy and those who received insulin therapy. We observed the lowest Lep expression in the PE group (6.3%). High level of HIF-1α expression was detected in T1DM (not planned) (30.44%) and PE (29.64%) as compared to the control group (11.62%). In T2DM and GDM insulin groups, the HIF-1α expression was significantly higher as compared to diet groups. CONCLUSION The obtained data show that DM and pre-eclampsia are associated with changes in angiogenic and metabolic placental factor expressions. The degree of changes depends on preconception care and the control of glycemia level during pregnancy.
Collapse
Affiliation(s)
- Roman V Kapustin
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, Department of Obstetrics, Division of Maternal-Fetal Medicine, St. Petersburg, Russia
- Department of Obstetrics, Gynecology and Reproduction, Faculty of Medicine, St. Petersburg State University, St Petersburg, Russia
| | - Ekaterina V Kopteeva
- Department of Obstetrics, Gynecology and Reproduction, Faculty of Medicine, St. Petersburg State University, St Petersburg, Russia
| | - Elena N Alekseenkova
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, Department of Obstetrics, Division of Maternal-Fetal Medicine, St. Petersburg, Russia
| | - Tatyana G Tral
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, Department of pathology, St. Petersburg, Russia
- Saint Petersburg State Pediatric Medical University of Health Ministry of Russia, Saint Petersburg, Russia
| | - Gulrukhsor Kh Tolibova
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, Department of pathology, St. Petersburg, Russia
- I.I. Mechnikov north-Western state Medical University, Saint Petersburg, Russia
| | - Olga N Arzhanova
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, Department of Obstetrics, Division of Maternal-Fetal Medicine, St. Petersburg, Russia
- Department of Obstetrics, Gynecology and Reproduction, Faculty of Medicine, St. Petersburg State University, St Petersburg, Russia
| |
Collapse
|
40
|
Gutaj P, Sibiak R, Jankowski M, Awdi K, Bryl R, Mozdziak P, Kempisty B, Wender-Ozegowska E. The Role of the Adipokines in the Most Common Gestational Complications. Int J Mol Sci 2020; 21:ijms21249408. [PMID: 33321877 PMCID: PMC7762997 DOI: 10.3390/ijms21249408] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/03/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
Adipocytokines are hormonally active molecules that are believed to play a key role in the regulation of crucial biological processes in the human body. Numerous experimental studies established significant alterations in the adipokine secretion patterns throughout pregnancy. The exact etiology of various gestational complications, such as gestational diabetes, preeclampsia, and fetal growth abnormalities, needs to be fully elucidated. The discovery of adipokines raised questions about their potential contribution to the molecular pathophysiology of those diseases. Multiple studies analyzed their local mRNA expression and circulating protein levels. However, most studies report conflicting results. Several adipokines such as leptin, resistin, irisin, apelin, chemerin, and omentin were proposed as potential novel early markers of heterogeneous gestational complications. The inclusion of the adipokines in the standard predictive multifactorial models could improve their prognostic values. Nonetheless, their independent diagnostic value is mostly insufficient to be implemented into standard clinical practice. Routine assessments of adipokine levels during pregnancy are not recommended in the management of both normal and complicated pregnancies. Based on the animal models (e.g., apelin and its receptors in the rodent preeclampsia models), future implementation of adipokines and their receptors as new therapeutic targets appears promising but requires further validation in humans.
Collapse
Affiliation(s)
- Paweł Gutaj
- Department of Reproduction, Chair of Obstetrics, Gynecology, and Gynecologic Oncology, Poznań University of Medical Sciences, 60-535 Poznan, Poland; (R.S.); (E.W.-O.)
- Correspondence: ; Tel.: +61-854-65-55
| | - Rafał Sibiak
- Department of Reproduction, Chair of Obstetrics, Gynecology, and Gynecologic Oncology, Poznań University of Medical Sciences, 60-535 Poznan, Poland; (R.S.); (E.W.-O.)
- Department of Histology and Embryology, Poznań University of Medical Sciences, 60-781 Poznan, Poland;
| | - Maurycy Jankowski
- Department of Anatomy, Poznań University of Medical Sciences, 60-781 Poznan, Poland; (M.J.); (R.B.)
| | - Karina Awdi
- Student’s Scientific Society, Poznan University of Medical Sciences, 60-806 Poznan, Poland;
| | - Rut Bryl
- Department of Anatomy, Poznań University of Medical Sciences, 60-781 Poznan, Poland; (M.J.); (R.B.)
| | - Paul Mozdziak
- Physiology Graduate Program, North Carolina State University, Raleigh, NC 27695-7608, USA;
| | - Bartosz Kempisty
- Department of Histology and Embryology, Poznań University of Medical Sciences, 60-781 Poznan, Poland;
- Department of Anatomy, Poznań University of Medical Sciences, 60-781 Poznan, Poland; (M.J.); (R.B.)
- Department of Obstetrics and Gynecology, University Hospital, Masaryk University, 625 00 Brno, Czech Republic
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, 87-100 Torun, Poland
| | - Ewa Wender-Ozegowska
- Department of Reproduction, Chair of Obstetrics, Gynecology, and Gynecologic Oncology, Poznań University of Medical Sciences, 60-535 Poznan, Poland; (R.S.); (E.W.-O.)
| |
Collapse
|
41
|
Beneventi F, Locatelli E, De Amici M, Cavagnoli C, Bellingeri C, De Maggio I, Ruspini B, Spinillo A. Maternal and fetal Leptin and interleukin 33 concentrations in pregnancy complicated by obesity and preeclampsia. J Matern Fetal Neonatal Med 2020; 33:3942-3948. [PMID: 30856361 DOI: 10.1080/14767058.2019.1593359] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Objectives: To evaluate maternal and fetal Leptin and IL33 concentrations in pregnancy complicated by obesity and preeclampsia.Study design: A case-control study including 35 subjects with obesity (18 normotensive and 17 preeclamptic) and 47 normal weight controls (42 normotensive and 5 preeclamptic).Main outcome measures: Leptin and IL33 concentrations in maternal serum during pregnancy and in cord blood; uterine artery and umbilical artery Doppler velocimetry.Results: Subjects with obesity who developed preeclampsia had higher first trimester maternal (41.5, interquartile range (IQR) = 15.7-65.1 ng/ml) Leptin concentrations compared to either normal weight with (25, IQR = 20.4-25.8 ng/ml) and without hypertension (14.26, IQR = 8.2-22.8) (p < .05) or normotensive subjects with obesity (30.3, IQR = 10.4-38.4) (p < .05). Subjects with obesity who developed preeclampsia (2.4, IQR = 1.7-3.2 pg/ml) or not (1.4, IQR = 0.8-2 pg/ml) had lower first trimester maternal IL33 levels when compared to controls without hypertension (4.8, IQR = 2.9-5.9 pg/ml) (p < .001). Cord blood Leptin and IL33 concentrations were significantly correlated to third trimester maternal concentrations (Spearman rho = 0.51, p < .001 and Spearman rho = 0.68, p < .001, respectively). Uterine artery pulsatility index (PI) were significantly and directly correlated with maternal Leptin levels (p < .002) and inversely and statistically correlated with maternal IL33 concentrations (p < .001).Conclusions: Compared to lean controls, pregnant subjects with obesity had higher serum Leptin and lower IL33 concentrations at first trimester and during pregnancy. This difference persisted also for those who later developed preeclampsia. The relationship between maternal serum levels of Leptin and IL33 with uterine artery Doppler pulsatility index strongly suggests a role of these two markers in early placentation.
Collapse
Affiliation(s)
- Fausta Beneventi
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Elena Locatelli
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Mara De Amici
- Department of Pediatrics, IRCCS Foundation Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Chiara Cavagnoli
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Camilla Bellingeri
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Irene De Maggio
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Beatrice Ruspini
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Arsenio Spinillo
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo and University of Pavia, Pavia, Italy
| |
Collapse
|
42
|
Galindo-Cáceres MA, Parra-Unda R, Murillo-Llanes J, Morgan-Ortiz F, Rendón-Maldonado JG, Osuna-Espinoza KY, Osuna-Ramírez I. Association of leptin receptor expression in placenta and peripheral blood mononuclear cell with maternal weight in birth outcomes. Cytokine 2020; 138:155362. [PMID: 33264748 DOI: 10.1016/j.cyto.2020.155362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/14/2020] [Accepted: 11/02/2020] [Indexed: 11/24/2022]
Abstract
INTRODUCTION The pregnancy period represents the most intense period of growth and development. Pre-pregnancy weight influences weight gain during pregnancy. Leptin is a hormone mainly derived from white adipose tissue, during pregnancy leptin is also produced by the placenta. It has been suggested that the effects of placental leptin on the mother may contribute to endocrine-mediated alterations in energy balance; a dysregulation in leptin levels or its receptors may lead to poor birth outcomes. Therefore, the main goal of the present study was to analyze the differences in birth outcomes by maternal weight with the expression level of leptin receptor in maternal peripheral blood mononuclear cell (PBMC) and placental tissue. METHODS Women with full-term gestation and its offspring were enrolled. Total RNA from maternal PBMC and placenta was obtained to perform the analysis of expression of the leptin receptor (LEPR) gene trough real-time PCR technique. Data were analyzed using one-way ANOVA or Mann-Whitney u test when applicable. Pearson correlation coefficient was used to determine the relationship between continuous variables (Stata v.13); p ≤ 0.05 was considered statistically significant. RESULTS No statistically significant differences were found between LEPR expression level and the BMI studied groups in maternal PBMC and placental tissue. Interaction between gestational weight gain (GWG) and LEPR in maternal PBMC explain in a 32% the variability of the newborn weight. CONCLUSIONS LEPR expression level in maternal PBMC correlates with newborn measurements independent from sex. GWG can affect fetal development by increasing fetal birth weight.
Collapse
Affiliation(s)
- M A Galindo-Cáceres
- Unidad de Investigaciones en Salud Pública, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Sinaloa, Mexico
| | - R Parra-Unda
- Unidad de Investigaciones en Salud Pública, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Sinaloa, Mexico
| | - J Murillo-Llanes
- Departamento de Investigación, Hospital de la Mujer de Culiacán, Servicios de Salud de Sinaloa, Culiacán, Sinaloa, Mexico
| | - F Morgan-Ortiz
- Ginecología y Obstetricia, Centro de Investigación y Docencia en Ciencias de la Salud, Hospital Civil de Culiacán, Culiacán, Sinaloa, Mexico
| | - J G Rendón-Maldonado
- Unidad de Investigaciones en Salud Pública, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Sinaloa, Mexico
| | - K Y Osuna-Espinoza
- Unidad Académica de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacán, Sinaloa, Mexico
| | - I Osuna-Ramírez
- Unidad de Investigaciones en Salud Pública, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Sinaloa, Mexico.
| |
Collapse
|
43
|
Zhang H, Xue L, Lv Y, Yu X, Zheng Y, Miao Z, Ding H. Integrated microarray analysis of key genes and a miRNA‑mRNA regulatory network of early‑onset preeclampsia. Mol Med Rep 2020; 22:4772-4782. [PMID: 33173953 PMCID: PMC7646902 DOI: 10.3892/mmr.2020.11551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/18/2020] [Indexed: 11/05/2022] Open
Abstract
Early‑onset preeclampsia (EOPE) is a serious threat to maternal and foetal health. The present study aimed to identify potential biomarkers and targets for the treatment of EOPE. Expression profiles of placenta from patients with EOPE and healthy controls (GSE103542, GSE74341 and GSE44711) were downloaded from the Gene Expression Omnibus database. Integrated analysis revealed 246 genes and 28 microRNAs (miRNAs) that were differentially expressed between patients with EOPE and healthy controls. Differentially expressed genes (DEGs) were primarily enriched in 'biological processes', such as 'cell adhesion', 'female pregnancy', 'extracellular matrix organization' and 'response to hypoxia'. Significant pathways associated with DEGs primarily included 'focal adhesion', 'ECM‑receptor interaction', 'PI3K‑Akt signaling' and 'ovarian steroidogenesis'. A Protein‑Protein Interaction network of DEGs was constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins online database, and epidermal growth factor receptor, collagen α‑1(I) chain, secreted phosphoprotein 1, leptin (LEP), collagen α‑2(I) chain (COL1A2), plasminogen activator inhibitor 1 (SERPINE1), Thy‑1 membrane glycoprotein, bone morphogenetic protein 4, vascular cell adhesion protein 1 and matrix metallopeptidase 1 were identified as hub genes. The alterations of hsa‑miR‑937, hsa‑miR‑148b*, hsa‑miR‑3907, hsa‑miR‑367*, COL1A2, LEP and SERPINE1 in placenta were validated using our local samples. Our research showed that the expression of hsa‑miR‑937, hsa‑miR‑1486*, hsa‑miR‑3907, hsa‑miR‑367* and hub genes in the placenta were closely associated with the pathophysiology of EOPE. hsa‑miR‑937, hsa‑miR‑1486*, hsa‑miR‑3907, hsa‑miR‑367* and hub genes could serve as biomarkers for diagnosis and as potential targets for the treatment of EOPE.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Internal Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Lu Xue
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu 210004, P.R. China
| | - Yan Lv
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu 210004, P.R. China
| | - Xiang Yu
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu 210004, P.R. China
| | - Yiwei Zheng
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu 210004, P.R. China
| | - Zhijing Miao
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu 210004, P.R. China
| | - Hongjuan Ding
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu 210004, P.R. China
| |
Collapse
|
44
|
Magalhães ESDS, Méio MDBB, Peixoto-Filho FM, Gonzalez S, da Costa ACC, Moreira MEL. Pregnancy-induced hypertension, preterm birth, and cord blood adipokine levels. Eur J Pediatr 2020; 179:1239-1246. [PMID: 32062709 DOI: 10.1007/s00431-020-03586-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 11/24/2022]
Abstract
Maternal hypertension may alter physiological parameters, dysregulating the release of hormones such as adipokines, thus influencing the fetal growth course. This study investigated whether hypertensive disorders of pregnancy alter cord blood adipokine levels and correlate these with anthropometric parameters in preterm infants. This is a prospective cohort study with pregnant women < 37-week gestation with and without hypertension and their offspring. Cord blood leptin, adiponectin, and ghrelin were analyzed by LUMINEX®. These adipokines were compared between the groups exposed or not to gestational hypertension using non-parametric statistical tests. The hypertensive pregnancies had significantly higher cord blood leptin (1.00 (IQR 0.67-1.20 ng/mL)) and adiponectin (18.52 (IQR 17.52-25.13 μg/mL)) levels than those without hypertension (0.07 (IQR 0.06-0.08 ng/mL) and 8.13 (IQR 6.50-8.68 μg/mL), respectively, p < 0.0001). The adipokine levels were higher in AGA and SGA infants in the exposed group for both moderate and late preterm. SGA had significantly higher ghrelin levels than the AGA infants. Ghrelin levels were negatively correlated with birth weight (r = - 0.613, p < 0.001), birth length (r = - 0.510, p < 0.001), head circumference (- 0.346, p < 0.002), and gestational age (r = - 0.612, p < 0.001).Conclusions: Our findings demonstrate an increase in adipokine levels in the cord blood of preterm newborn infants exposed to maternal hypertension. What is Known: • Clinical evidence suggests that concentration of the serum adipokines may be affected by risk of hypertension in both adults and pregnant women. • Maternal profile as hypertension alters intrauterine environment and could affect the function of fetal metabolism, impairing fetal growth. What is New: • Gestational hypertension modifies the adipokine profile, with higher rates already present at birth in cord blood samples. • Within the hypertensive group and stratifying for gestation age, ghrelin concentrations were higher in SGA newborns, both in the moderate and late preterm, compared with AGA newborns.
Collapse
Affiliation(s)
- Elizabeth Soares da Silva Magalhães
- Clinical Research Unit, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Instituto Fernandes Figueira, Fundação Oswaldo Cruz, Av. Rui Barbosa, 716, Flamengo, Rio de Janeiro, RJ, 22250-020, Brazil.
| | - Maria Dalva Barbosa Baker Méio
- Clinical Research Unit, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Instituto Fernandes Figueira, Fundação Oswaldo Cruz, Av. Rui Barbosa, 716, Flamengo, Rio de Janeiro, RJ, 22250-020, Brazil
| | - Fernando Maia Peixoto-Filho
- Department of Obstetrics, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Sayonara Gonzalez
- Laboratory of Genomic Medicine, Department of Medical Genetics, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Ana Carolina Carioca da Costa
- Clinical Research Unit, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Instituto Fernandes Figueira, Fundação Oswaldo Cruz, Av. Rui Barbosa, 716, Flamengo, Rio de Janeiro, RJ, 22250-020, Brazil
| | - Maria Elisabeth Lopes Moreira
- Clinical Research Unit, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Instituto Fernandes Figueira, Fundação Oswaldo Cruz, Av. Rui Barbosa, 716, Flamengo, Rio de Janeiro, RJ, 22250-020, Brazil
| |
Collapse
|
45
|
Robineau-Charette P, Grynspan D, Benton SJ, Gaudet J, Cox BJ, Vanderhyden BC, Bainbridge SA. Fibrinogen-Like Protein 2-Associated Transcriptional and Histopathological Features of Immunological Preeclampsia. Hypertension 2020; 76:910-921. [PMID: 32713274 PMCID: PMC7418930 DOI: 10.1161/hypertensionaha.120.14807] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Preeclampsia is a multifactorial hypertensive disorder of pregnancy, with variable presentation in both maternal and fetal factors, such that no treatment or marker is currently universal to all cases. Here, we demonstrate that the prothrombinase and immunomodulatory secreted factor FGL-2 (fibrinogen-like protein 2) is differentially expressed across previously characterized gene expression clusters containing clinically relevant disease subtypes. FGL2 is low in a cluster consistent with the traditional paradigm of the pathology of preeclampsia (canonical preeclampsia) and high in a cluster exhibiting evidence of immune activation (immunological preeclampsia). We show that it is part of an immunoregulatory gene module integral to the transcriptional profile and placental pathology specific to immunological preeclampsia. We determine that FGL2 associates positively with chronic inflammation lesions of the placenta while associating negatively with maternal vascular malperfusion lesions. The transcriptional profiles of maternal vascular malperfusion lesions show downregulation of FGL2 and upregulation of previously investigated preeclampsia biomarkers, such as FLT1 (Fms Related Receptor Tyrosine Kinase 1) and ENG (endoglin). Conversely, the profiles of chronic inflammation lesions show an interesting downregulation of these genes, but an upregulation of FGL2 and of FGL2-correlated immunoregulatory genes, suggesting it is upregulated downstream of major inflammatory mediators such as TNF (tumor necrosis factor)-α and IFN (interferon)-γ, hallmarks of the immunological preeclampsia subtype. This work, overall, demonstrates that FGL-2 expression levels in the term placenta reflect the unique pathophysiology that leads to immunological preeclampsia, leading to its potential as a subtype-specific biomarker.
Collapse
Affiliation(s)
- Pascale Robineau-Charette
- From the Department of Cellular and Molecular Medicine (P.R.-C., S.J.B, B.C.V., S.A.B.), University of Ottawa, ON, Canada.,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada (P.R.-C., B.C.V.)
| | - David Grynspan
- Department of Pathology and Laboratory Medicine (D.G.), University of Ottawa, ON, Canada
| | - Samantha J Benton
- From the Department of Cellular and Molecular Medicine (P.R.-C., S.J.B, B.C.V., S.A.B.), University of Ottawa, ON, Canada
| | - Jeremiah Gaudet
- Faculty of Medicine and Interdisciplinary School of Health Sciences, Faculty of Health Sciences (J.G., S.A.B.), University of Ottawa, ON, Canada
| | - Brian J Cox
- Department of Physiology (B.J.C.), Faculty of Medicine, University of Toronto, ON, Canada.,Department of Obstetrics and Gynecology (B.J.C.), Faculty of Medicine, University of Toronto, ON, Canada
| | - Barbara C Vanderhyden
- From the Department of Cellular and Molecular Medicine (P.R.-C., S.J.B, B.C.V., S.A.B.), University of Ottawa, ON, Canada.,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada (P.R.-C., B.C.V.)
| | - Shannon A Bainbridge
- From the Department of Cellular and Molecular Medicine (P.R.-C., S.J.B, B.C.V., S.A.B.), University of Ottawa, ON, Canada.,Faculty of Medicine and Interdisciplinary School of Health Sciences, Faculty of Health Sciences (J.G., S.A.B.), University of Ottawa, ON, Canada
| |
Collapse
|
46
|
Gyselaers W. Preeclampsia Is a Syndrome with a Cascade of Pathophysiologic Events. J Clin Med 2020; 9:jcm9072245. [PMID: 32679789 PMCID: PMC7409017 DOI: 10.3390/jcm9072245] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/05/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
This review integrates the currently available information on the molecular, cellular, and systemic mechanisms involved in the pathophysiology of preeclampsia. It highlights that the growth, protection, and promotion of the conceptus requires the modulation of an intact maternal immune system, communication between the mother and fetus, and adaptation of the maternal organic functions. A malfunction in any of these factors, on either side, will result in a failure of the cascade of events required for the normal course of pregnancy. Maladaptive processes, initially aiming to protect the conceptus, fail to anticipate the gradually increasing cardiovascular volume load during the course of pregnancy. As a result, multiple organ dysfunctions install progressively and eventually reach a state where mother and/or fetus are at risk of severe morbidity or even mortality, and where the termination of pregnancy becomes the least harmful solution. The helicopter view on pathophysiologic processes associated with preeclampsia, as presented in this paper, illustrates that the etiology of preeclampsia cannot be reduced to one single mechanism, but is to be considered a cascade of consecutive events, fundamentally not unique to pregnancy.
Collapse
Affiliation(s)
- Wilfried Gyselaers
- Department Obstetrics, Ziekenhuis Oost Limburg, B3600 Genk, Belgium; ; Tel.: +32-89-306420
- Department Physiology, Hasselt University, B3590 Diepenbeek, Belgium
| |
Collapse
|
47
|
Plowden TC, Zarek SM, Rafique S, Sjaarda LA, Schisterman EF, Silver RM, Yeung EH, Radin R, Hinkle SN, Galai N, Mumford SL. Preconception leptin levels and pregnancy outcomes: A prospective cohort study. Obes Sci Pract 2020; 6:181-188. [PMID: 32313676 PMCID: PMC7156817 DOI: 10.1002/osp4.399] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 11/27/2019] [Accepted: 11/30/2019] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Obesity has become a major, worldwide public health issue and is associated with a greater risk of adverse pregnancy outcomes. Leptin, a hormone produced by adipocytes, is elevated in individuals with obesity and may mediate the association between obesity and pregnancy outcomes. Though leptin levels during pregnancy have been associated with pregnancy outcomes, less is understood regarding preconception levels. Therefore, the objective of this study was to evaluate associations between preconception leptin levels and adverse pregnancy outcomes. METHODS This was a prospective cohort study nested within a large randomized controlled trial conducted at four medical centres in the United States. A total of 1078 women completed the parent study; this analysis involved women who became pregnant during that study (n = 776). Patients were healthy women, ages 18 to 40, attempting to conceive, with 1 to 2 prior pregnancy losses. Participants were followed for less than or equal to 6 cycles while trying to conceive and throughout pregnancy if they conceived. Preconception leptin concentrations were measured in serum collected at baseline then categorized by tertiles (using the lowest as reference group). Weighted log-binomial regression estimated risk ratios (RR) and 95% confidence intervals (CIs) for pregnancy loss, preterm delivery (PTD), gestational diabetes (GDM), and hypertensive disorders in pregnancy, adjusting for age, waist-to-hip ratio (WHR), and body mass index (BMI). RESULTS The mean (SD) BMI in this cohort was 25.4 ± 6.0. GDM (RR 18.37; 95% CI, 2.39-141.55) and hypertensive disorders of pregnancy (RR 2.35; 95% CI, 1.20-4.61) risks were higher among women in the high tertile after adjusting for age and WHR. The associated risk persisted when adjusting for BMI for GDM but was attenuated for hypertensive disorders in pregnancy. Leptin levels were not associated with risk of pregnancy loss or PTD. CONCLUSIONS Women with higher baseline preconception leptin levels had a higher likelihood of experiencing some adverse pregnancy outcomes including GDM and hypertensive disorders of pregnancy. These findings warrant further evaluation, especially in light of the association between leptin and obesity.
Collapse
Affiliation(s)
- Torie C. Plowden
- Epidemiology Branch, Division of Intramural Population Health ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentBethesdaMarylandUSA
- Program in Reproductive and Adult EndocrinologyEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaMarylandUSA
| | - Shvetha M. Zarek
- Epidemiology Branch, Division of Intramural Population Health ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentBethesdaMarylandUSA
- Program in Reproductive and Adult EndocrinologyEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaMarylandUSA
| | - Saima Rafique
- Department of Obstetrics and GynecologyHoward University HospitalWashingtonDCUSA
| | - Lindsey A. Sjaarda
- Epidemiology Branch, Division of Intramural Population Health ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentBethesdaMarylandUSA
| | - Enrique F. Schisterman
- Epidemiology Branch, Division of Intramural Population Health ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentBethesdaMarylandUSA
| | - Robert M. Silver
- Department of Obstetrics and GynecologyUniversity of Utah Health Sciences CenterSalt Lake CityUtahUSA
| | - Edwina H. Yeung
- Epidemiology Branch, Division of Intramural Population Health ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentBethesdaMarylandUSA
| | - Rose Radin
- Epidemiology Branch, Division of Intramural Population Health ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentBethesdaMarylandUSA
| | - Stefanie N. Hinkle
- Epidemiology Branch, Division of Intramural Population Health ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentBethesdaMarylandUSA
| | - Noya Galai
- Department of StatisticsUniversity of HaifaHaifaIsrael
| | - Sunni L. Mumford
- Epidemiology Branch, Division of Intramural Population Health ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentBethesdaMarylandUSA
| |
Collapse
|
48
|
Hao S, You J, Chen L, Zhao H, Huang Y, Zheng L, Tian L, Maric I, Liu X, Li T, Bianco YK, Winn VD, Aghaeepour N, Gaudilliere B, Angst MS, Zhou X, Li YM, Mo L, Wong RJ, Shaw GM, Stevenson DK, Cohen HJ, Mcelhinney DB, Sylvester KG, Ling XB. Changes in pregnancy-related serum biomarkers early in gestation are associated with later development of preeclampsia. PLoS One 2020; 15:e0230000. [PMID: 32126118 PMCID: PMC7053753 DOI: 10.1371/journal.pone.0230000] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/19/2020] [Indexed: 12/19/2022] Open
Abstract
Background Placental protein expression plays a crucial role during pregnancy. We hypothesized that: (1) circulating levels of pregnancy-associated, placenta-related proteins throughout gestation reflect the temporal progression of the uncomplicated, full-term pregnancy, and can effectively estimate gestational ages (GAs); and (2) preeclampsia (PE) is associated with disruptions in these protein levels early in gestation; and can identify impending PE. We also compared gestational profiles of proteins in the human and mouse, using pregnant heme oxygenase-1 (HO-1) heterozygote (Het) mice, a mouse model reflecting PE-like symptoms. Methods Serum levels of placenta-related proteins–leptin (LEP), chorionic somatomammotropin hormone like 1 (CSHL1), elabela (ELA), activin A, soluble fms-like tyrosine kinase 1 (sFlt-1), and placental growth factor (PlGF)–were quantified by ELISA in blood serially collected throughout human pregnancies (20 normal subjects with 66 samples, and 20 subjects who developed PE with 61 samples). Multivariate analysis was performed to estimate the GA in normal pregnancy. Mean-squared errors of GA estimations were used to identify impending PE. The human protein profiles were then compared with those in the pregnant HO-1 Het mice. Results An elastic net-based gestational dating model was developed (R2 = 0.76) and validated (R2 = 0.61) using serum levels of the 6 proteins measured at various GAs from women with normal uncomplicated pregnancies. In women who developed PE, the model was not (R2 = -0.17) associated with GA. Deviations from the model estimations were observed in women who developed PE (P = 0.01). The model developed with 5 proteins (ELA excluded) performed similarly from sera from normal human (R2 = 0.68) and WT mouse (R2 = 0.85) pregnancies. Disruptions of this model were observed in both human PE-associated (R2 = 0.27) and mouse HO-1 Het (R2 = 0.30) pregnancies. LEP outperformed sFlt-1 and PlGF in differentiating impending PE at early human and late mouse GAs. Conclusions Serum placenta-related protein profiles are temporally regulated throughout normal pregnancies and significantly disrupted in women who develop PE. LEP changes earlier than the well-established biomarkers (sFlt-1 and PlGF). There may be evidence of a causative action of HO-1 deficiency in LEP upregulation in a PE-like murine model.
Collapse
Affiliation(s)
- Shiying Hao
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children’s Hospital, Palo Alto, CA, United States of America
| | - Jin You
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Lin Chen
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Hui Zhao
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Yujuan Huang
- Department of Emergency, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Le Zheng
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children’s Hospital, Palo Alto, CA, United States of America
| | - Lu Tian
- Department of Health Research and Policy, Stanford University, Stanford, CA, United States of America
| | - Ivana Maric
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Xin Liu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Tian Li
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Ylayaly K. Bianco
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Virginia D. Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Martin S. Angst
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Xin Zhou
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China
| | - Yu-Ming Li
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China
| | - Lihong Mo
- Department of Obstetrics and Gynecology, University of California San Francisco-Fresno, Fresno, CA, United States of America
| | - Ronald J. Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Gary M. Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America
| | - David K. Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Harvey J. Cohen
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Doff B. Mcelhinney
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children’s Hospital, Palo Alto, CA, United States of America
| | - Karl G. Sylvester
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Xuefeng B. Ling
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children’s Hospital, Palo Alto, CA, United States of America
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
- * E-mail:
| |
Collapse
|
49
|
Adipokines and Endothelium Dysfunction Markers in Pregnant Women with Gestational Hypertension. Int J Hypertens 2019; 2019:7541846. [PMID: 31737362 PMCID: PMC6815564 DOI: 10.1155/2019/7541846] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 08/07/2019] [Accepted: 09/13/2019] [Indexed: 12/17/2022] Open
Abstract
Objective The aim of the study was to evaluate the levels of adipokines such as adiponectin and leptin as well as soluble intercellular adhesion molecule-1 (sICAM-1) and endogenous NOS inhibitor-asymmetric dimethylarginine (ADMA), as the endothelium dysfunction markers in pregnant women with gestational hypertension (GH). Patients and Methods Adiponectin, leptin, sICAM-1, and ADMA concentrations were measured in a group of 34 patients with GH and in 32 healthy pregnant women between the 24th and 34th week of gestation with ELISA tests. Results The patients with GH compared with healthy ones were characterized by significantly higher BMI (28.09 ± 7.90 vs. 22.34 ± 4.21 kg/m2, p=0.016) and higher concentrations of leptin (45.89 ± 35.91 vs. 24.09 ± 24.40 ng/mL, p=0.006). sICAM-1 levels were also higher in the GH group but without the statistical significance (264.51 ± 50.99 vs. 232.56 ± 43.3 ng/ml, p=0.057). There were no significant differences between groups in adiponectin (8.79 ± 8.67 vs. 7.90 ± 3.71 μg/mL, p=0.46, NS) and ADMA (0.57 ± 0.26 vs. 0.60 ± 0.24 μmol/L, p=0.68, NS) levels. The significant correlation between leptin levels and BMI value was observed only in patients with GH (R = 0.56, p=0.02). Conclusions The higher levels of leptin in pregnant women with gestational hypertension may be suggestive of the role of leptin in GH development. As the patients in the GH group had higher BMI, hyperleptinemia may link obesity with gestational hypertension. The significance of leptin as the predictive marker of GH development could be implied. It could be postulated that the higher levels of sICAM-1 in the GH patients, although not statistically significant, could reflect some impairment of the endothelium function occurring in GH regardless of BMI. The comparable adiponectin levels in GH and healthy pregnant patients and the lack of its correlation with BMI may indicate the occurrence of a protective mechanism in pregnancy maintaining its concentration and preserving from the consequences of the decrease in its levels in overweight and obese patients. Since ADMA levels were similar in GH and healthy pregnant women, ADMA seems not to be involved in GH pathogenesis, suggesting that NO synthesis is not impaired in this pregnancy complication. As the data on the gestational hypertension pathogenesis and its correlations with adipokines and markers of the endothelium dysfunction are limited, further studies on this issue are warranted.
Collapse
|
50
|
Lackner HK, Papousek I, Schmid-Zalaudek K, Cervar-Zivkovic M, Kolovetsiou-Kreiner V, Nonn O, Lucovnik M, Pfniß I, Moertl MG. Disturbed Cardiorespiratory Adaptation in Preeclampsia: Return to Normal Stress Regulation Shortly after Delivery? Int J Mol Sci 2019; 20:ijms20133149. [PMID: 31252672 PMCID: PMC6651868 DOI: 10.3390/ijms20133149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 12/30/2022] Open
Abstract
Women with pregnancies complicated by preeclampsia appear to be at increased risk of metabolic and vascular diseases in later life. Previous research has also indicated disturbed cardiorespiratory adaptation during pregnancy. The aim of this study was to follow up on the physiological stress response in preeclampsia several weeks postpartum. A standardized laboratory test was used to illustrate potential deviations in the physiological stress responding to mildly stressful events of the kind and intensity in which they regularly occur in further everyday life after pregnancy. Fifteen to seventeen weeks postpartum, 35 women previously affected by preeclampsia (19 mild, 16 severe preeclampsia), 38 women after uncomplicated pregnancies, and 51 age-matched healthy controls were exposed to a self-relevant stressor in a standardized stress-reactivity protocol. Reactivity of blood pressure, heart rate, stroke index, and systemic vascular resistance index as well as baroreceptor sensitivity were analyzed. In addition, the mutual adjustment of blood pressure, heart rate, and respiration, partitioned for influences of the sympathetic and the parasympathetic branches of the autonomic nervous system, were quantified by determining their phase synchronization. Findings indicated moderately elevated blood pressure levels in the nonpathological range, reduced stroke volume, and elevated systemic vascular resistance in women previously affected by preeclampsia. Despite these moderate abnormalities, at the time of testing, women with previous preeclampsia did not differ from the other groups in their physiological response patterns to acute stress. Furthermore, no differences between early, preterm, and term preeclampsia or mild and severe preeclampsia were observed at the time of testing. The findings suggest that the overall cardiovascular responses to moderate stressors return to normal in women who experience a pregnancy with preeclampsia a few weeks after delivery, while the operating point of the arterial baroreflex is readjusted to a higher pressure. Yet, their regulation mechanisms may remain different.
Collapse
Affiliation(s)
- Helmut K Lackner
- Division of Physiology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria.
| | - Ilona Papousek
- Department of Psychology, Biological Psychology Unit, University of Graz, 8010 Graz, Austria.
| | - Karin Schmid-Zalaudek
- Division of Physiology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria.
| | - Mila Cervar-Zivkovic
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria.
| | | | - Olivia Nonn
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria.
| | - Miha Lucovnik
- Department of Perinatology, Division of Obstetrics and Gynecology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia.
| | - Isabella Pfniß
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria.
| | - Manfred G Moertl
- Department of Obstetrics and Gynecology, Clinical Center, 9020 Klagenfurt, Austria.
| |
Collapse
|