1
|
Smith S, Smith J, Jones K, Castillo A, Wiemann N, Howard-Cunningham A, Cunningham M. Placental ischemia during pregnancy induces hypertension, cerebral inflammation, and oxidative stress in dams postpartum. Hypertens Pregnancy 2025; 44:2454597. [PMID: 39885618 PMCID: PMC11849403 DOI: 10.1080/10641955.2025.2454597] [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: 10/16/2024] [Accepted: 01/07/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Preeclampsia (PE) is characterized as de novo hypertension (HTN) with end-organ damage, especially in the brain. PE is hypothesized to be caused by placental ischemia. PE affects ~5-8% of USA pregnancies and increases the risk for HTN and cerebrovascular diseases (CVD) later in life. We hypothesize that blood pressure (BP), cerebral oxidative stress, and cerebral inflammation will increase in postpartum (PP) placental ischemic dams. METHODS Placental ischemia was induced in pregnant Sprague Dawley dams, utilizing reduced uterine perfusion pressure (RUPP) surgery. At 6 weeks PP (~3 human years), BP was measured via carotid catheterization, and cerebral oxidative stress and inflammation were assessed via ELISAs, biochemical assays, and Western blots. RESULTS BP, cerebral pro-inflammatory cytokines (TNF-α and IL-6), and GFAP (a marker of astrocyte activity) were increased in PP RUPP dams. Cerebral hydrogen peroxide (H2O2) was also increased in PP RUPP dams, and had a strong correlation with PP RUPP BP, proinflammatory cytokines (TNF- α and IL-6), and GFAP astrocyte activation. CONCLUSION PP RUPP dams have increased BP, cerebral oxidative stress, and cerebral inflammation at 6 weeks postpartum. These changes in cerebral inflammation and oxidative stress may contribute to the pathology and development of HTN and CVDs in postpartum dams.
Collapse
Affiliation(s)
- Savanna Smith
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX
| | - Jonna Smith
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX
| | - Kylie Jones
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX
| | - Angie Castillo
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX
| | - Natalia Wiemann
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX
| | | | - Mark Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX
| |
Collapse
|
2
|
Ma RC, Chow EW, Ng NY, Sugiyama T, Deng Y, Tam WH, Lau SL, Poon LC, Zhang C, Sriram U. Diabetes and women's health. J Diabetes Investig 2025. [PMID: 40492908 DOI: 10.1111/jdi.70095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2025] [Revised: 05/20/2025] [Accepted: 05/23/2025] [Indexed: 06/12/2025] Open
Abstract
There is increasing interest in the implementation of precision medicine in diabetes. Gender is an important determinant of health, and there are numerous characteristics of diabetes in women that can be taken into account for the implementation of precision medicine in diabetes. Furthermore, better appreciation of the unique clinical characteristics of diabetes in women, especially in relation to the lifecourse perspective of a woman's life, would greatly enhance public health efforts to address diabetes and its associated risk factors, and to improve the lives of women and the next generation. These include the unique challenges and opportunities linked with polycystic ovary syndrome, gestational diabetes, other pregnancy complications including preeclampsia, and the potential impact of these diagnoses in the offspring of affected women. In this review article, we will summarize some of these key aspects of how diabetes intersects with women's health, and how a holistic, integrative, and lifecourse approach to diabetes and women's health can have a significant impact well beyond just the benefits of achieving better glucose control for the women living with diabetes, but more importantly, toward improved prevention and control of noncommunicable diseases on a population level.
Collapse
Affiliation(s)
- Ronald Cw Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Edith Wk Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
- Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Noel Yh Ng
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Takashi Sugiyama
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Yuzhi Deng
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wing Hung Tam
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong SAR, China
- CUHK Medical Centre, Shatin, Hong Kong
| | - So Ling Lau
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Liona C Poon
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Cuilin Zhang
- Global Centre for Asian Women's Health (GLOW), National University of Singapore, Singapore, Singapore
| | - Usha Sriram
- TAG Centre Department of Diabetes and Endocrinology, Voluntary Health Services, Chennai, India
| |
Collapse
|
3
|
Zhu S, Chen S, Ge Y, Zhou F, Su K, Xu C, Wu Y, Huang H. High-fat diet induces pre-eclampsia through dampening cell-autonomous C3 in trophoblasts. Commun Biol 2025; 8:879. [PMID: 40481230 PMCID: PMC12144106 DOI: 10.1038/s42003-025-08298-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
Hyperlipidemia contributes to low-grade inflammation and abnormal immune response, which is putatively involved in the development of pre-eclampsia (PE). As components of innate immune system, complements play a critical role in regulating inflammation. However, how cell-autonomous complement changes and works in PE remains elusive. In the current study, we established L-NAME-induced mice to manifest PE-like symptoms. In presence of high-fat diet (HFD) feeding, the PE-like symptoms were considerably aggravated, as well as down-regulated complement C3 in HFD/L-NAME mice trophoblasts. To explore the effect of C3 in PE development, we generated C3 overexpression and knockdown cell (Swan.71C3 and Swan.71ΔC3) based on Swan.71, a trophoblast cell line. We found that Swan.71C3 cells display promoted proliferation, migration and invasion capability and less secretion of anti-angiogenetic cytokines, while Swan.71ΔC3 showed highly-activated NLRP3 inflammasome and pyroptosis, which was also noted in HFD/L-NAME placentas, highlighting the contributing role of inflammation to PE. Indeed, pro-inflammatory cytokines were increased in placentas from HFD/L-NAME mice. The similar trends of C3 in trophoblast from severe PE patients supported the contribution role of C3 to PE pathogenesis. Thus, our study provides evidence that cell-autonomous complement C3 regulates NLRP3 inflammasome activation upon HFD exposure, affecting trophoblast cell function in PE development.
Collapse
Affiliation(s)
- Simeng Zhu
- Department of Cardiology, Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siyue Chen
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Yingzhou Ge
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Fangyue Zhou
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Kaizhen Su
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Congfeng Xu
- Department of Cardiology, Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Yanting Wu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
- International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
- State Key Laboratory of Cardiology, Shanghai, China.
| |
Collapse
|
4
|
Shabani F, Jodeiri A, Mohammad-Alizadeh-Charandabi S, Abbasalizadeh F, Tanha J, Mirghafourvand M. Developing and validating an artificial intelligence-based application for predicting some pregnancy outcomes: a multi-phase study protocol. Reprod Health 2025; 22:99. [PMID: 40481447 PMCID: PMC12144753 DOI: 10.1186/s12978-025-02048-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 05/25/2025] [Indexed: 06/11/2025] Open
Abstract
Background Pregnancy complications such as preterm birth, low birth weight, gestational diabetes mellitus, preeclampsia, and intrauterine growth restriction significantly affect both maternal and neonatal health outcomes. Early identification of high-risk pregnancies is essential for timely interventions; however, traditional predictive models often lack accuracy. This study aims to develop and validate an AI-based application to improve risk assessment and clinical decision-making regarding pregnancy outcomes through a multi-phase approach. Methods This study comprises three phases. In Phase 1, retrospective case-control data will be collected from medical records, including Mother and Infant System (IMaN), Hospital Information System (HIS), and archived records of women who gave birth at Al-Zahra and Taleghani Educational and Medical Centers in Tabriz between 2022 and 2024. In Phase 2, an artificial intelligence model will be developed using machine learning algorithms such as Random Forest, XGBoost, Support Vector Machines (SVM), and neural networks, followed by model training, validation, and integration into a user-friendly application. Phase 3 will focus on a prospective cohort study of pregnant women attending clinics after 22 weeks of gestation, evaluating the AI model’s predictive performance through metrics like AUROC (area under the receiver operating characteristic curve), sensitivity, specificity, and predictive values, along with real-time data collection. Content validity will be determined through expert reviews. Discussion This study protocol presents a multi-phase approach to developing and validating an AI-based application for predicting pregnancy outcomes. By integrating retrospective data analysis, machine learning, and prospective validation, the study aims to improve early risk detection and maternal care. If successful, this application could support personalized obstetric decision-making. This study aims to develop and validate an artificial intelligence (AI)-based tool to predict pregnancy complications, including preterm birth, low birth weight, gestational diabetes, intrauterine growth restriction, and preeclampsia. The research will be conducted in three phases. First, past medical records from two hospitals will be analysed to identify key risk factors. Next, a machine learning model will be developed and integrated into a user-friendly application. Finally, the tool will be tested on a group of pregnant women to assess its accuracy in predicting adverse pregnancy outcomes. By leveraging AI, this study seeks to enhance early risk detection, enabling healthcare providers to implement timely preventive measures and improve maternal and neonatal health outcomes. If successful, this AI-based application could serve as a valuable resource in maternity care, assisting midwives and doctors in delivering personalized care and reducing complications. The findings could also advance the use of AI technology in obstetric practice, improving decision-making and optimizing healthcare resources.
Collapse
Affiliation(s)
- Fatemeh Shabani
- Midwifery Department, Faculty of Nursing and Midwifery, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Jodeiri
- Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Fatemeh Abbasalizadeh
- Department of Obstetrics and Gynecology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Tanha
- Faculty of Electrical and Computer Engineering, University of Tabriz, Tabriz, Iran
| | - Mojgan Mirghafourvand
- Social Determinants of Health Research Center, Faculty of Nursing and Midwifery, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
5
|
Xu Z, Li EH, Liu J, Zhang YJ, Xiao R, Chen XZ, Zhong ZH, Tang XJ, Fu LJ, Zhang H, Bao MH, Qi HB, Chen GL, Ding YB. Postpartum hemorrhage emerges as a key outcome of maternal SARS-CoV-2 omicron variant infection surge across pregnancy trimesters. J Infect Public Health 2025; 18:102733. [PMID: 40073664 DOI: 10.1016/j.jiph.2025.102733] [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: 07/25/2024] [Revised: 02/27/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Despite evidence showing changes in disease severity with the virus's evolution and vaccination efforts, the link between maternal, perinatal, and neonatal outcomes and SARS-CoV-2 infections during different pregnancy trimesters remains unclear, especially with the BA.5 and BF.7 Omicron subvariant surge in China in December 2022. This study investigates the correlation between maternal, perinatal, and neonatal outcomes and SARS-CoV-2 infection across various pregnancy trimesters. METHODS This prospective cohort study was conducted at two hospitals in southwest China, examining the clinical records and infection status of 2158 pregnant women registered between January 1, 2022, and September 30, 2023. Initially shielded from COVID-19, the population later experienced a significant infection surge. A comparative analysis evaluated maternal, perinatal, and neonatal outcomes between infected and uninfected subjects. Primary outcomes included pregnancy complications and premature births, while secondary outcomes encompassed cesarean sections, delivery complications, and neonatal outcomes. RESULTS Pregnant women infected with SARS-CoV-2 had higher incidence of placenta increta/percreta and postpartum hemorrhage compared to uninfected women. First trimester infections were associated with a lower incidence of intrahepatic cholestasis of pregnancy [aOR = 0.29, 95 % CI 0.13-0.63] but a higher incidence of preterm birth [aOR = 2.16, 95 % CI 1.25-3.71]. Third trimester infections increased the risk of postpartum hemorrhage [aOR = 2.74, 95 % CI 1.21-6.18]. CONCLUSION SARS-CoV-2 infection during pregnancy is linked to increased incidence of placenta increta/percreta and postpartum hemorrhage. First trimester infections are associated with higher incidence of premature birth and lower incidence of intrahepatic cholestasis of pregnancy, while third trimester infections are linked to higher incidence of postpartum hemorrhage.
Collapse
Affiliation(s)
- Zhou Xu
- Department of Obstetrics and Gynecology, Sichuan Jinxin Xinan Women and Children's Hospital, Chengdu, Sichuan 610011, China
| | - Er-Han Li
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China; Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Jia Liu
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Yong-Jia Zhang
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China; Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Rui Xiao
- Department of Obstetrics and Gynecology, Sichuan Jinxin Xinan Women and Children's Hospital, Chengdu, Sichuan 610011, China
| | - Xin-Zhen Chen
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Zhao-Hui Zhong
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Xiao-Jun Tang
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Li-Juan Fu
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China; Department of Pharmacology, Academician Workstation, Changsha Medical University, Changsha 410219, China
| | - Hua Zhang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016 China
| | - Mei-Hua Bao
- Department of Pharmacology, Academician Workstation, Changsha Medical University, Changsha 410219, China
| | - Hong-Bo Qi
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China.
| | - Gong-Li Chen
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China.
| | - Yu-Bin Ding
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China; Department of Pharmacology, Academician Workstation, Changsha Medical University, Changsha 410219, China.
| |
Collapse
|
6
|
Mahan VL. Heme oxygenase/carbon monoxide system affects the placenta and preeclampsia. Med Gas Res 2025; 15:276-287. [PMID: 39829164 PMCID: PMC11918472 DOI: 10.4103/mgr.medgasres-d-24-00081] [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: 09/06/2024] [Revised: 10/17/2024] [Accepted: 11/25/2024] [Indexed: 01/22/2025] Open
Abstract
Preeclampsia affects 2% to 8% of pregnancies worldwide and results in significantly high maternal and perinatal morbidity and mortality, with delivery being the only definitive treatment. It is not a single disorder, but rather a manifestation of an insult(s) to the uteroplacental unit -whether maternal, fetal, and/or placental. Multiple etiologies have been implicated, including uteroplacental ischemia, maternal infection and/or inflammation, maternal obesity, sleep disorders, hydatidiform mole, maternal intestinal dysbiosis, autoimmune disorders, fetal diseases, breakdown of maternal-fetal immune tolerance, placental aging, and endocrine disorders. Early- and late-onset preeclampsia are associated with different etiologies: early-onset preeclampsia develops because of poor placentation, while late-onset preeclampsia occurs in women with latent maternal endothelial dysfunction. In preeclamptic placentas, acquired, genetic, and immune risk factors may result in impaired trophoblast invasion and spiral artery remodeling, which affects uteroplacental perfusion. The resulting placental hypoxia affects the heme oxygenase system-a known stress response pathway affected by hypoxia that is important during normal pregnancy and may offer a therapeutic approach in preeclampsia. This review will address the effect of the heme oxygenase/carbon monoxide system on the placenta and preeclampsia.
Collapse
Affiliation(s)
- Vicki L. Mahan
- Department of Surgery, Queen Elizabeth Central Hospital, Blantyre, Malawi
- Drexel University Medical School, Philadelphia, PA, USA
| |
Collapse
|
7
|
Xu Q, Xing L, Zhang T, Liu G. Development and validation of a preeclampsia prediction model for the first and second trimester pregnancy based on medical history. BMC Pregnancy Childbirth 2025; 25:616. [PMID: 40426100 PMCID: PMC12107935 DOI: 10.1186/s12884-025-07733-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
OBJECTIVE The study aimed to identify the risk factors of preeclampsia (PE) and establish a novel prediction model. STUDY DESIGN A retrospective, single-center analysis was conducted using clinical data from 5099 pregnant women who gave birth at Peking University People's Hospital between June 2015 and December 2020 who had placental growth factor (PIGF) levels records at 13-20 + 6 gestation weeks. The participants were randomly divided into a training set (70%, n = 3569) and a validation set (30%, n = 1030), between which the consistency was checked, and the analysis was performed according to whether PE occurred during pregnancy. Factors with univariate logistic analysis outcome of p < 0.2 were incorporated into the multivariate logistic regression analysis model, then variable selection by stepwise regression with AIC as the criterion was executed to finally identify the variables used for modeling. The model's discriminative ability was assessed using the receiver operating characteristic (ROC) curve, and its calibration was evaluated through calibration curves and Hosmer-Lemesow test. In addition, decision curve analysis (DCA) was used for clinical net benefit appraisal. RESULTS Logistic regression analysis identified nine risk factors for PE, including: maternal age (OR = 1.072, 95%CI = 1.025-1.120), parity(OR = 0.718,95%CI = 0.470-1.060), pre-pregnancy BMI (OR = 2.842,95%CI = 1.957-4.106), family hypertension history (OR = 3.604,95%CI = 2.433-5.264), pregestational diabetes mellitus(PGDM) (OR = 8.399, 95%CI = 4.138-15.883), pregnancy complicating nephropathy (OR = 7.931, 95% CI = 2.584-20.258),pregnancy complicating immune system disorders (OR = 3.134, 95% CI = 1.624-5.525), mean arterial pressure(MAP) at 11-13 + 6 gestational weeks (OR = 1.098, 95% CI = 1.078-1.119) and PIGF (OR = 0.647, 95% CI = 0.448-0.927) at 13-20 + 6 gestational weeks (P < 0.05). The restricted spline regression analysis (RCS) analysis results showed that PIGF and the risk of PE presented an approximately "L-shaped" relationship, with the risk of PE rising sharply with the decrease of PIGF when PIGF < 90 pg/ml, and little change with the increase of PIGF when PIGF > 90 pg/ml. A risk prediction model for PE during the first and second trimester was constructed based on the above selected 11 factors. The area under the ROC curve (AUC) for the model was 0.781(95%CI = 0.709-0.853), and the sensitivity and specificity at the optimal cut-off value (threshold probability) were 0.571 and 0.879 respectively. Chi-square of 9.616 and P value of 0.293 from Hosmer-Lemeshow test indicated that the model was well calibrated. Finally, the model showed good clinical net benefits in the threshold range of 0.03-0.3. CONCLUSION The incidence of PE was associated with maternal age, pre-pregnancy weight and BMI, family hypertension history, PGDM, pregnancy complicating nephropathy, gestational complicating immune system disorders, blood pressure (systolic, diastolic, mean arterial pressure) at 11-13 + 6 gestational weeks, and PIGF at 13-20 + 6 gestational weeks. When PIGF < 90 pg/ml at 13-20 + 6 gestational week, the risk of PE increased significantly with the reduction of PIGF. The nomogram based on the above results was simpler and more practical in clinical application for PE predicting during the first and second trimester, and may provide an important reference for doctors and patients.
Collapse
Affiliation(s)
- Qi Xu
- Obstetrics and Gynaecology Department, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, P.R. China
| | - Lili Xing
- Obstetrics and Gynaecology Department, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, P.R. China
| | - Ting Zhang
- Obstetrics and Gynaecology Department, Obstetrics and Gynaecology Department, Ordos Obstetrics and Gynecology Hospital, No.9 Wansheng Ring Road, Dongsheng District, Ordos City, Inner Mongolia Autonomous Region, P.R. China
| | - Guoli Liu
- Obstetrics and Gynaecology Department, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, P.R. China.
| |
Collapse
|
8
|
Hua Q, Yang F, Zhou Y, Shi F, You X, Guo J, Li L. Predictive Models Using Machine Learning to Identify Fetal Growth Restriction in Patients With Preeclampsia: Development and Evaluation Study. J Med Internet Res 2025; 27:e70068. [PMID: 40424611 DOI: 10.2196/70068] [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: 12/29/2024] [Revised: 03/20/2025] [Accepted: 04/15/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Fetal growth restriction (FGR) is a common complication of preeclampsia. FGR in patients with preeclampsia increases the risk of neonatal-perinatal mortality and morbidity. However, previous prediction methods for FGR are class-biased or clinically unexplainable, which makes it difficult to apply to clinical practice, leading to a relative delay in intervention and a lack of effective treatments. OBJECTIVE The study aims to develop an auxiliary diagnostic model based on machine learning (ML) to predict the occurrence of FGR in patients with preeclampsia. METHODS This study used a retrospective case-control approach to analyze 38 features, including the basic medical history and peripheral blood laboratory test results of pregnant patients with preeclampsia, either complicated or not complicated by FGR. ML models were constructed to evaluate the predictive value of maternal parameter changes on preeclampsia combined with FGR. Multiple algorithms were tested, including logistic regression, light gradient boosting, random forest (RF), extreme gradient boosting, multilayer perceptron, naive Bayes, and support vector machine. The model performance was identified by the area under the curve (AUC) and other evaluation indexes. The Shapley additive explanations (SHAP) method was adopted to rank the feature importance and explain the final model for clinical application. RESULTS The RF model performed best in discriminative ability among the 7 ML models. After reducing features according to importance rank, an explainable final RF model was established with 9 features, including urinary protein quantification, gestational week of delivery, umbilical artery systolic-to-diastolic ratio, amniotic fluid index, triglyceride, D-dimer, weight, height, and maximum systolic pressure. The model could accurately predict FGR for 513 patients with preeclampsia (149 with FGR and 364 without FGR) in the training and testing dataset (AUC 0.83, SD 0.03) using 5-fold cross-validation, which was closely validated for 103 patients with preeclampsia (n=45 with FGR and n=58 without FGR) in an external dataset (AUC 0.82, SD 0.048). On the whole, urinary protein quantification, umbilical artery systolic-to-diastolic ratio, and gestational week of delivery exhibited the highest contributions to the model performance (c=0.45, 0.34, and 0.33) based on SHAP analysis. For specific individual patients, SHAP results reveal the protective and risk factors to develop FGR for interpreting the model's clinical significance. Finally, the model has been translated into a convenient web page tool to facilitate its use in clinical settings. CONCLUSIONS The study successfully developed a model that accurately predicts FGR development in patients with preeclampsia. The SHAP method captures highly relevant risk factors for model interpretation, alleviating concerns about the "black box" problem of ML techniques.
Collapse
Affiliation(s)
- Qing Hua
- Department of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Fengchun Yang
- Institute of Medical Information, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Yadan Zhou
- Department of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Fenglian Shi
- Department of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Xiaoyan You
- Department of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Jing Guo
- Department of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Li Li
- Department of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Jia H, Liu S, Wang W, He P, Zhao F, Xu X. Microplastic exposure induces preeclampsia-like symptoms via HIF-1α/TFRC-mediated ferroptosis in placental trophoblast cells. Toxicology 2025; 516:154197. [PMID: 40414414 DOI: 10.1016/j.tox.2025.154197] [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: 03/20/2025] [Revised: 05/12/2025] [Accepted: 05/18/2025] [Indexed: 05/27/2025]
Abstract
Microplastic (MP) pollution is an emerging environmental concern with potential health risks, yet its impact on pregnancy remains largely unexplored. This study investigated the effects of polystyrene microplastic (PS-MP) exposure on placental function and its role in preeclampsia (PE) pathogenesis. Pregnant rats were exposed to PS-MP, which induced PE-like symptoms including elevated blood pressure, increased proteinuria, and altered expression of angiogenic factors. Transcriptomic and molecular analyses revealed PS-MP triggered ferroptosis in placental trophoblast cells by activating the HIF-1α/TFRC axis, resulting in iron overload and oxidative stress. PS-MP exposure impaired trophoblast migration, invasion, and angiogenesis; these effects were ameliorated by ferroptosis inhibition. These findings identified PS-MP-induced ferroptosis as a critical mechanism underlying placental dysfunction, highlighting PS-MP as a potential environmental risk factor for PE. Understanding the impact of MP on pregnancy provides crucial insights into their reproductive toxicity and underscores the need for further research on mitigating their effects.
Collapse
Affiliation(s)
- Haoyi Jia
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Siyu Liu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Wenhao Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Pengyuan He
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Fujun Zhao
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Department of Andrology, Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| | - Xianming Xu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| |
Collapse
|
10
|
Onishi K, Kawakita T. Association of Gestational Weight Gain with Adverse Pregnancy Outcomes in Individuals with Obesity. Am J Perinatol 2025. [PMID: 40311624 DOI: 10.1055/a-2597-8542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
This study aimed to determine optimal gestational weight gain (GWG) considering adverse infant and maternal outcomes among individuals with obesity, with detailed classification for body mass index (BMI) of 40 kg/m2 or higher.This study was a population-based retrospective cohort study, using U.S. birth certificate data from 2017 to 2021. We included nulliparous individuals with singleton pregnancies delivering live births between 370/7 and 416/7 weeks' gestation, excluding those with pregestational diabetes or hypertension, gestational diabetes, preeclampsia, major fetal anomalies, or chromosomal disorders. The study focused on three main outcomes: primary cesarean delivery, small for gestational age (SGA), and large for gestational age (LGA). GWG was evaluated in 2-kg increments, from weight loss >8 kg to gains ≥28 kg. Prepregnancy BMI was stratified into four categories: BMI of 30 to <35, 35 to <40, 40 to <50, and ≥50 kg/m2. Odds ratios and absolute risk reduction were used to identify GWG ranges with balanced risks for three outcomes within each BMI category.Among 1,677,968 individuals with obesity, increased GWG was associated with higher absolute risks of cesarean delivery and LGA and lower risk of SGA across all BMI categories. Optimal GWG ranges varied by prepregnancy BMI: >12 to ≤14 kg for BMI of 30 to <35 kg/m2; >10 to ≤12 kg for BMI of 35 to <40 kg/m2; >6 to ≤10 kg for BMI of 40 to <50 kg/m2; >0 to ≤8 kg for BMI of 50 kg/m2 or higher.We identified higher upper and lower GWG limits for individuals with BMI of 30 to 50 kg/m2, and lower limits for those with BMI ≥50 kg/m2, compared with the guidelines recommended by the U.S. National Academy of Medicine. These findings suggest the need to tailor GWG recommendations based on the severity of obesity. · Optimal GWG varies by obesity class, requiring tailored guidelines.. · Those with BMI ≥50 kg/m may need lower target for gestational gain than current recommendation.. · The optimal GWG range could be changed based on the outcomes of interest..
Collapse
Affiliation(s)
- Kazuma Onishi
- Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, Virginia
- Department of Reproductive Medicine, Asada Ladies Clinic, Nagoya, Aichi, Japan
| | - Tetsuya Kawakita
- Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, Virginia
| |
Collapse
|
11
|
Zhou Y, Chen Y, Li L, Lin L. Inhibition of hsa_circ_0003314 contributes to trophoblast cell migration and invasion and inhibits pyroptosis in preeclampsia. Histochem Cell Biol 2025; 163:52. [PMID: 40377691 DOI: 10.1007/s00418-025-02384-3] [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] [Accepted: 04/28/2025] [Indexed: 05/18/2025]
Abstract
Inflammation is a key contributor to the development of preeclampsia. Recent studies suggest that circular RNAs (circRNAs) may serve as potential therapeutic targets for this disease, though their specific functions remain incompletely understood. In this study, we investigated the role of hsa_circ_0003314 in preeclampsia pathogenesis. The interaction between hsa_circ_0003314 and microRNA (miR)-1827 was validated using RNA pull-down and luciferase reporter assays, while the binding of miR-1827 to the 3'-UTR of caspase-5 was confirmed by RNA immunoprecipitation and luciferase reporter assays. Pyroptotic cells were quantified by flow cytometry based on the percentage of caspase-1/propidium iodide (PI) double-positive cells. Enzyme-linked immunosorbent assay (ELISA) was performed to measure interleukin (IL)-1β concentrations in the culture supernatant. The migration and invasion abilities of HTR-8/SVneo cells were evaluated using Transwell assays. We found that hsa_circ_0003314 expression was upregulated in HTR-8/SVneo cells subjected to hypoxia/reoxygenation (H/R) treatment. Silencing hsa_circ_0003314 enhanced cell migration, invasion, and epithelial-mesenchymal transition (EMT), while reducing the expression of pyroptosis-related proteins, GSDMD-N and HMGB1. The proportion of pyroptotic cells was significantly decreased upon hsa_circ_0003314 knockdown in H/R-treated cells. Mechanistically, hsa_circ_0003314 functions as a molecular sponge for miR-1827, thereby regulating caspase-5 expression. Notably, caspase-5 overexpression rescued the effects of hsa_circ_0003314 knockdown, restoring pyroptosis markers and suppressing the enhanced migratory and invasive behavior of HTR-8/SVneo cells. In conclusion, silencing hsa_circ_0003314 promotes migration, invasion, and EMT in H/R-treated HTR-8/SVneo cells by inhibiting caspase-5-mediated pyroptosis through the sequestration of miR-1827. These findings identify hsa_circ_0003314 as a promising therapeutic target in the treatment of preeclampsia.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Obstetrics and Gynecology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, No. 134 Dong Street, Fuzhou, 350001, Fujian, China
| | - Yuqing Chen
- Department of Obstetrics and Gynecology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, No. 134 Dong Street, Fuzhou, 350001, Fujian, China
| | - Lihua Li
- Department of Obstetrics and Gynecology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, No. 134 Dong Street, Fuzhou, 350001, Fujian, China
| | - Lizhen Lin
- Department of Obstetrics and Gynecology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, No. 134 Dong Street, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
12
|
Karabay G, Seyhanli Z, Filiz AA, Cakir BT, Aktemur G, Sucu S, Vanli Tonyali N, Akin F, Karabay U, Yilmaz ZV. Fetal Aortic Isthmus Doppler Evaluation in Pre-Eclampsia Patients. JOURNAL OF CLINICAL ULTRASOUND : JCU 2025. [PMID: 40370296 DOI: 10.1002/jcu.24079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 03/31/2025] [Accepted: 04/28/2025] [Indexed: 05/16/2025]
Abstract
PURPOSE This study aimed to evaluate the role of fetal aortic isthmus (AoI) Doppler parameters in predicting adverse perinatal outcomes in pre-eclampsia patients. METHODS This prospective study included 60 pre-eclampsia patients (divided into early-onset [EOPE] and late-onset [LOPE] groups) and 74 controls from Ankara Etlik City Hospital. Doppler parameters-such as aortic isthmus flow index (IFI), systolic/diastolic ratio (S/D), pulsatility index (PI), and resistive index (RI)-were collected, and their associations with adverse perinatal outcomes were analyzed using ROC analysis. RESULTS In the EOPE group, gestational age, birth weight, and APGAR scores were significantly lower compared to the LOPE and control groups (p < 0.001). The EOPE group also showed significantly higher rates of NICU (neonatal intensive care unit) admission, preterm birth, respiratory distress syndrome, and need for mechanical ventilation (p < 0.001). The cut-off values for IFI (> 1.13) and AoI S/D (> 7.42) were found to be significant predictors of adverse perinatal outcomes. CONCLUSION AoI Doppler parameters may aid in predicting adverse neonatal outcomes in pre-eclampsia patients. The values of IFI, S/D, PI, and RI show potential as useful tools in clinical management, especially in monitoring high-risk pregnancies.
Collapse
Affiliation(s)
- Gulsan Karabay
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Zeynep Seyhanli
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Ahmet Arif Filiz
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Betul Tokgoz Cakir
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Gizem Aktemur
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Sadun Sucu
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Nazan Vanli Tonyali
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Furkan Akin
- Department of Obstetrics and Gynecology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Umut Karabay
- Department of Internal Medicine, Gulhane Training and Research Hospital, Ankara, Turkey
| | - Zehra Vural Yilmaz
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| |
Collapse
|
13
|
Pilar Fernández-Figares Vicioso M, Riutord Sbert P, López-González ÁA, Ramírez-Manent JI, Del Barrio Fernández JL, Herrero MTV. Risk of Insulin Resistance: Comparison of the Commerce vs. Industry Sector and Associated Variables. Diseases 2025; 13:150. [PMID: 40422582 DOI: 10.3390/diseases13050150] [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: 04/07/2025] [Revised: 05/04/2025] [Accepted: 05/12/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Insulin resistance (IR) is a key metabolic alteration that precedes type 2 diabetes and is closely linked to obesity and lifestyle factors. Occupational context may influence IR risk through variations in physical activity, diet, and socioeconomic determinants. OBJECTIVE To compare the risk of insulin resistance between workers in the commerce and industry sectors and identify associated sociodemographic and lifestyle factors, in order to improve their occupational health. METHODS This cross-sectional study analyzed data from 56,856 Spanish workers, assessing four IR-related indices: Triglyceride-Glucose Index (TyG), TyG-BMI (Triglyceride-Glucose Body Mass Index), Metabolic Score for Insulin Resistance (METS-IR), and the Single-Point Insulin Sensitivity Estimator (SPISE-IR). The analysis was stratified by sex and sector (commerce vs. industry) and included assessments of age, education level, physical activity, adherence to the Mediterranean diet, and smoking status. Multinomial logistic regressions were performed to determine the factors associated with high IR scores. RESULTS Across all IR indicators, industry workers-particularly men-presented higher mean values and greater prevalence of high-risk scores compared to those in commerce. Women showed lower values overall but also reflected sector-based differences. In both sexes, non-physical activity, non-adherence to the Mediterranean diet, and smoking were consistently associated with higher IR risk. Males exhibited significantly higher odds of elevated TyG (OR = 2.59, 95% CI: 2.41-2.78), while physical inactivity and poor diet emerged as the most powerful modifiable predictors across all scales (e.g., OR = 10.45 for TyG, OR = 12.33 for TyG-BMI). Industry sector was independently associated with higher odds of insulin resistance compared to commerce. CONCLUSIONS Insulin resistance is more prevalent among industrial workers, especially men and those with unhealthy lifestyles. Occupational health strategies should target sector-specific risk profiles, emphasizing physical activity and dietary interventions.
Collapse
Affiliation(s)
| | - Pere Riutord Sbert
- ADEMA-Health Group of IUNICS, University of Balearic Islands, 07122 Palma, Spain
| | - Ángel Arturo López-González
- Obesity and Metabolic Syndrome Group, Spanish Association of Specialists in Occupational Medicine, 28012 Madrid, Spain
- ADEMA-Health Group of IUNICS, University of Balearic Islands, 07122 Palma, Spain
| | - José Ignacio Ramírez-Manent
- ADEMA-Health Group of IUNICS, University of Balearic Islands, 07122 Palma, Spain
- Health Research Institute of the Balearic Islands (IDISBA), 07120 Palma, Spain
- Faculty of Medicine, University of Balearic Islands, 07122 Palma, Spain
| | | | - María Teófila Vicente Herrero
- Obesity and Metabolic Syndrome Group, Spanish Association of Specialists in Occupational Medicine, 28012 Madrid, Spain
- ADEMA-Health Group of IUNICS, University of Balearic Islands, 07122 Palma, Spain
| |
Collapse
|
14
|
Starodubtseva N, Poluektova A, Tokareva A, Kukaev E, Avdeeva A, Rimskaya E, Khodzayeva Z. Proteome-Based Maternal Plasma and Serum Biomarkers for Preeclampsia: A Systematic Review and Meta-Analysis. Life (Basel) 2025; 15:776. [PMID: 40430203 PMCID: PMC12113278 DOI: 10.3390/life15050776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 04/29/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Proteomics has emerged as a transformative tool in biomedical research, enabling comprehensive characterization of protein profiles in complex biological systems. In preeclampsia (PE) research, quantitative proteomic analyses of plasma and serum have revealed critical insights into disease mechanisms and potential biomarkers. Through a systematic review of 17 studies (2009-2024), we identified 561 differentially expressed plasma/serum proteins (p < 0.05) in PE patients versus healthy controls, with 122 proteins consistently replicated across ≥2 independent studies. Stratified analysis by clinical subtype (early-vs. late-onset PE) demonstrated both concordant and divergent protein expression patterns, reflecting heterogeneity in PE pathophysiology, methodological variations (e.g., sample processing, proteomic platforms), and differences between discovery-phase and targeted validation studies. The trimester-specific biomarker panels proposed here offer a framework for future large-scale, multicenter validation. By integrating advanced proteomic technologies with standardized preanalytical and analytical protocols, these findings advance opportunities for early prediction (first-trimester biomarker signatures); mechanistic insight (complement system involvement); and personalized management (subtype-specific therapeutic targets). This work underscores the potential of proteomics to reshape PE research, from molecular discovery to clinical translation, ultimately improving outcomes for this leading cause of maternal and perinatal morbidity.
Collapse
Affiliation(s)
- Natalia Starodubtseva
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
| | - Alina Poluektova
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
| | - Alisa Tokareva
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
| | - Evgenii Kukaev
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
- V.L. Talrose Institute for Energy Problems of Chemical Physics, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Anna Avdeeva
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
| | - Elena Rimskaya
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
- Lebedev Physical Institute, 119991 Moscow, Russia
| | - Zulfiya Khodzayeva
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
| |
Collapse
|
15
|
Mensah NA, Fassett MJ, Peltier MR, Shi J, Chiu VY, Khadkha N, Getahun D. Joint Effect of Body Mass Index and Obstructive Sleep Apnea on Preeclampsia Risk. Am J Perinatol 2025. [PMID: 40194541 DOI: 10.1055/a-2576-4082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Preeclampsia remains one of the leading causes of perinatal mortality worldwide. Little is known about the modifiable risk factors that can be identified and addressed early in pregnancy to reduce the risk of preeclampsia and its associated adverse outcomes. We sought to determine if there is a synergistic effect of prepregnancy body-mass index and obstructive sleep apnea (OSA) on the risk of preeclampsia.We conducted a retrospective cohort study of singleton pregnancies delivered in Kaiser Permanente Southern California hospitals between January 1, 2010, and December 31, 2020 (n = 342,349). Preeclampsia and sleep apnea were ascertained using clinical diagnosis codes. Body mass index (BMI) in kg/m2 measured during prenatal care visits was categorized as normal (18.5-24.9), overweight (25-29.9), and obese (≥30). Multivariable logistic regression was used to estimate adjusted relative risks (aRR) and 95% confidence intervals (CI).Compared with normal weight in pregnancy, overweight (aRR : 1.6; 95% CI: 1.5, 1.7) and obese BMI (aRR: 2.5; 95% CI: 2.4, 2.6) were associated with an increased risk of preeclampsia. Independent of prepregnancy body-mass index, a pregnancy with OSA was associated with an increased risk of preeclampsia (aRR: 2.2; 95% CI: 1.8, 2.6). Compared with normal weight without the diagnosis of OSA in a pregnancy, overweight (aRR: 4.6; 95% CI: 2.9, 7.4) and obese BMI (aRR: 3.8; 95% CI: 3.2, 4.6) with the diagnosis of OSA were associated with an increased risk of preeclampsia.OSA and elevated body-mass index have an independent and additive relationship with preeclampsia. Overweight women at risk of preeclampsia should be advised of a higher likelihood of developing preeclampsia when both conditions occur together and may benefit from close monitoring and early interventions for these modifiable risk factors. · There is a dose-dependent association between BMI and the risk of preeclampsia.. · Coexistent obesity and OSA resulted in a stronger risk for preeclampsia.. · The combined effect of obesity and OSA on preeclampsia risk is additive rather than synergistic..
Collapse
Affiliation(s)
- Nana A Mensah
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, California
- Department of Public Health, Brigham Young University, Provo, Utah
| | - Michael J Fassett
- Department of Obstetrics & Gynecology, Kaiser Permanente West Los Angeles Medical Center, California
- Department of Clinical Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, California
| | - Morgan R Peltier
- Department of Psychiatry and Behavioral Health, Jersey Shore University Medical Center, Neptune, New Jersey
- Department of Psychiatry and Behavioral Health, Hackensack Meridian School of Medicine, Nutley, New Jersey
| | - Jiaxiao Shi
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, California
| | - Vicki Y Chiu
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, California
| | - Nehaa Khadkha
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, California
| | - Darios Getahun
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, California
- Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, California
| |
Collapse
|
16
|
Chatzakis C, Papavasiliou D, Mansukhani T, Nicolaides KH, Charakida M. Maternal vascular-placental axis in the third trimester in women with gestational diabetes mellitus, hypertensive disorders, and unaffected pregnancies. Am J Obstet Gynecol 2025; 232:489.e1-489.e11. [PMID: 39218286 DOI: 10.1016/j.ajog.2024.08.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 08/10/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Hypertensive disorders of pregnancy and gestational diabetes mellitus are characterized by vascular dysfunction and are associated with long term cardiovascular risks. OBJECTIVE This study aimed to compare different markers of maternal vascular function in women with gestational diabetes mellitus, preeclampsia, or gestational hypertension and in women whose pregnancies were unaffected by these complications and to assess the association between maternal vascular function and markers of placental perfusion and maternal vascular-placental axis in 4 groups of women. STUDY DESIGN This was a prospective observational study of women who had routine hospital visits at 35 0/7 to 36 6/7 weeks of gestation at King's College Hospital, London, United Kingdom. The routine hospital visit included recording of maternal demographic characteristics and medical history, ultrasound examination for fetal anatomy and growth, Doppler studies of the uterine arteries and ophthalmic arteries, carotid-femoral pulse wave velocity measurements, estimation of the augmentation index and total peripheral resistance, and measurements of serum placental growth factor and soluble fms-like tyrosine kinase 1. Linear regression analysis was performed for the outcomes of uterine artery pulsatility index multiple of the median, placental growth factor multiple of the median, and soluble fms-like tyrosine kinase 1 multiple of the median. The ophthalmic artery peak systolic velocity ratio, pulse wave velocity, augmentation index, and total peripheral vascular resistance were assessed as potential predictors. This analysis was performed on all women and separately in the different groups. RESULTS The study population of 6502 women included 614 (9.4%) with gestational diabetes mellitus, 140 (2.1%) who subsequently developed preeclampsia, and 129 (2.0%) who developed gestational hypertension. Women with gestational diabetes mellitus had increased pulse wave velocity compared with those with pregnancies unaffected by gestational diabetes mellitus, preeclampsia, or gestational hypertension. Women with preeclampsia or gestational hypertension had lower placental growth factor multiple of the median and higher uterine artery pulsatility index multiple of the median, soluble fms-like tyrosine kinase 1 multiple of the median, augmentation index, pulse wave velocity, total peripheral resistance, and ophthalmic artery peak systolic velocity ratio than those with unaffected pregnancies. In women with unaffected pregnancies, the ophthalmic artery peak systolic velocity ratio was predictive of the uterine artery pulsatility index multiple of the median, and ophthalmic artery peak systolic velocity ratio, augmentation index, total peripheral resistance, and pulse wave velocity were predictive of the placental growth factor multiple of the median and the soluble fms-like tyrosine kinase 1 multiple of the median. In women with gestational diabetes mellitus, the ophthalmic artery peak systolic velocity ratio was predictive of the uterine artery pulsatility index multiple of the median; the ophthalmic artery peak systolic velocity ratio, total peripheral resistance, and pulse wave velocity were predictive of the placental growth factor multiple of the median; and total peripheral resistance was predictive of the soluble fms-like tyrosine kinase 1 multiple of the median. In women with preeclampsia, the ophthalmic artery peak systolic velocity ratio was predictive of the uterine artery pulsatility index multiple of the median, placental growth factor multiple of the median, and soluble fms-like tyrosine kinase 1 multiple of the median. In women unaffected by gestational diabetes mellitus, preeclampsia, or gestational hypertension, the ophthalmic artery peak systolic velocity ratio was predictive of the uterine artery pulsatility index multiple of the median, and the augmentation index, total peripheral resistance, pulse wave velocity, and the ophthalmic artery peak systolic velocity ratio were predictive of the placental growth factor multiple of the median and the soluble fms-like tyrosine kinase 1 multiple of the median. CONCLUSION In the third trimester of pregnancy, women with preeclampsia, gestational hypertension, and gestational diabetes mellitus present with increased arterial stiffness. In addition, women diagnosed with hypertensive complications showed increased peripheral vascular resistance. The ophthalmic artery peak systolic velocity ratio provided predictive information for placental perfusion and function in all pregnant women, whereas vascular indices were more informative for placental function in women with unaffected pregnancies and those with gestational diabetes mellitus than in those with preeclampsia or gestational hypertension. Our data suggest that vascular assessment in women during pregnancy not only may provide information about maternal vascular health but also can be used to provide information about individual risk factors for placental insufficiency. The selection of the vascular index will have to be tailored according to the maternal profile and pregnancy complication.
Collapse
Affiliation(s)
- Christos Chatzakis
- Second Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - Dimitra Papavasiliou
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom
| | - Tanvi Mansukhani
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom
| | - Kypros H Nicolaides
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom.
| | - Marietta Charakida
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom; School of Biomedical Engineering and Imaging Sciences, King's College Hospital, London, United Kingdom
| |
Collapse
|
17
|
de Oliveira AA, Spaans F, Graton ME, Stokes A, Kirschenman R, Quon A, Cooke CLM, Davidge ST. Aspirin Improves Uterine Artery Function in Hypercholesterolemic Preeclampsia. Hypertension 2025; 82:859-871. [PMID: 39936305 DOI: 10.1161/hypertensionaha.124.24435] [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: 12/06/2024] [Accepted: 01/31/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Excessive hypercholesterolemia in pregnancy increases the risk of preeclampsia (HC-PE), though the mechanisms remain unclear. We recently showed that uterine artery function is impaired in HC-PE pregnancies via activation of the TLR4 (toll-like receptor 4)/PGHS1 (prostaglandin H synthase 1) pathway. Low-dose aspirin lowers preeclampsia risk in high-risk pregnancies by inhibiting PGHS1, but its effects in HC-PE pregnancies are not known. Moreover, oxidized low-density lipoprotein (oxLDL) levels rise in HC-PE, potentially activating TLR4 and LOX-1 (lectin-like oxLDL receptor-1; scavenger receptor linked to vascular dysfunction in preeclampsia). However, whether this occurs in HC-PE is not known. METHODS Sprague Dawley rats received a control or high-cholesterol diet (to induce HC-PE) from gestational day 6 to 20, with placebo or low-dose aspirin (1.5 mg/kg daily) given from gestational day 10 to 20. On gestational day 20, pregnancy outcomes and uterine artery function were assessed. RESULTS Uterine artery blood flow velocity and placental weights were higher in HC-PE placebo-treated dams versus controls, but these were reduced by low-dose aspirin. Endothelium-dependent vasodilation was impaired in the uterine arteries of the HC-PE placebo group versus controls and was corrected by low-dose aspirin. Ex vivo inhibition of TLR4, PGHS1, or LOX-1 also normalized endothelium-dependent vasodilation in the HC-PE placebo-treated dams. Exposure to oxLDL in the bath (modeling a secondary hit) further impaired endothelium-dependent vasodilation in the uterine arteries of the HC-PE placebo group, partially via TLR4 and LOX-1, which was prevented by low-dose aspirin. CONCLUSIONS Low-dose aspirin improved uterine artery endothelial function in HC-PE pregnancies; likely by suppressing the TLR4/LOX-1/PGHS1 pathway.
Collapse
Affiliation(s)
- Amanda A de Oliveira
- Department of Obstetrics and Gynecology (A.A.d.O., F.S., M.E.G., R.K., A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (A.A.d.O., F.S., M.E.G., A.S., R.K., A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Floor Spaans
- Department of Obstetrics and Gynecology (A.A.d.O., F.S., M.E.G., R.K., A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (A.A.d.O., F.S., M.E.G., A.S., R.K., A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Murilo E Graton
- Department of Obstetrics and Gynecology (A.A.d.O., F.S., M.E.G., R.K., A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (A.A.d.O., F.S., M.E.G., A.S., R.K., A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Angie Stokes
- Department of Biological Sciences (A.S.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (A.A.d.O., F.S., M.E.G., A.S., R.K., A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Raven Kirschenman
- Department of Obstetrics and Gynecology (A.A.d.O., F.S., M.E.G., R.K., A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (A.A.d.O., F.S., M.E.G., A.S., R.K., A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Anita Quon
- Department of Obstetrics and Gynecology (A.A.d.O., F.S., M.E.G., R.K., A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (A.A.d.O., F.S., M.E.G., A.S., R.K., A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Christy-Lynn M Cooke
- Department of Obstetrics and Gynecology (A.A.d.O., F.S., M.E.G., R.K., A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (A.A.d.O., F.S., M.E.G., A.S., R.K., A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Sandra T Davidge
- Department of Obstetrics and Gynecology (A.A.d.O., F.S., M.E.G., R.K., A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Department of Physiology (S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (A.A.d.O., F.S., M.E.G., A.S., R.K., A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| |
Collapse
|
18
|
Wu H, Zhao L. Circ_0015382 Regulates the miR-942-5p/NDRG1 Axis to Suppress Trophoblast Cell Functions. Reprod Sci 2025; 32:1491-1501. [PMID: 39354288 DOI: 10.1007/s43032-024-01685-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/18/2024] [Indexed: 10/03/2024]
Abstract
Circular RNAs (circRNAs) are non-coding RNAs that exert vital function in many human diseases, including preeclampsia (PE). Circ_0015382 is considered to be a key regulator of PE progression, so more molecular mechanisms need to be further studied. Real-time quantitative PCR was used to detect the mRNA levels of circ_0015382, miR-942-5p, and N-myc downstream regulated 1 (NDRG1). Western blot analysis was conducted to assess the protein levels. MTT and EdU assays were used to assess cell proliferation. Cell invasion was analyzed by transwell assay. Tube formation assay was conducted to detect cell angiogenesis. Dual-luciferase reporter assay and RNA immunoprecipitation were used to analyze the target relationship between miR-942-5p and circ_0015382 or NDRG1. Our data showed that circ_0015382 and NDRG1 were up-regulated, while miR-942-5p was down-regulated in the placental tissues of PE patients. Trophoblast cell proliferation, invasion, and angiogenesis were promoted by knockdown of circ_0015382. Circ_0015382 could sponge miR-942-5p, and NDRG1 was a target of miR-942-5p. MiR-942-5p inhibitor could reverse the promoting effects of si-circ_0015382 on trophoblast cell functions. Besides, the enhancing effects of miR-942-5p mimic on trophoblast cell proliferation, invasion, and angiogenesis could be eliminated by NDRG1 overexpression. In conclusion, our data showed that circ_0015382 inhibited trophoblast cell proliferation, invasion, and angiogenesis through regulating miR-942-5p/NDRG1 axis, providing a new mechanism for the regulation of PE progression.
Collapse
Affiliation(s)
- Haiyan Wu
- Obstetrical Department, Northwest Women's and Children's Hospital, No.1616 Yanxiang Road, Qujiang New District, Xi'an, Shaanxi, 710061, China
| | - Lijuan Zhao
- Obstetrical Department, Northwest Women's and Children's Hospital, No.1616 Yanxiang Road, Qujiang New District, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
19
|
Ramos A, Youssef L, Molina P, Martinez‐Sanchez J, Moreno‐Castaño AB, Blasco M, De Moner B, Tortajada M, Camacho M, Borrell M, Crovetto F, Pino M, Escolar G, Carreras E, Gratacos E, Diaz‐Ricart M, Palomo M, Crispi F. Endothelial damage and complement dysregulation in fetuses from pregnancies complicated by preeclampsia. Acta Obstet Gynecol Scand 2025; 104:829-838. [PMID: 40007223 PMCID: PMC11981108 DOI: 10.1111/aogs.15072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 01/19/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025]
Abstract
INTRODUCTION Our objective was to evaluate the endothelial function profile and complement system in fetuses from preeclamptic pregnancies using ex vivo and in vitro approaches. MATERIAL AND METHODS A total of 66 singleton pregnancies were prospectively recruited comprising 34 cases of preeclampsia and 32 normotensive pregnancies matched for baseline characteristics. In the ex vivo approach, soluble tumor necrosis factor-a receptor 1 (sTNFR1), vascular cell adhesion molecule-1 (sVCAM-1), intercellular adhesion molecule-1 (sICAM-1), Von Willebrand factor (sVWF), terminal complement complex (sC5b-9), Factor H, complement component C3a and Factor Bb were analyzed in fetal cord blood samples. In the in vitro model, vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), Von Willebrand factor (VWF), vascular endothelial cadherin (VE-Cadherin), endothelial nitric oxide synthase (eNOS), reactive oxygen species (ROS) and C5b-9 deposits were evaluated on endothelial cells in culture exposed to fetal sera or plasma. RESULTS Increased sVCAM-1, sICAM- l and decreased Factor H and Factor Bb concentrations were detected in preeclampsia fetuses as compared to fetuses from normotensive mothers (509.4 ± 28 vs. 378.4 ± 34.3 ng/mL, 161.1 ± 11.9 vs. 114.8 ± 6.8, 199.6 ± 18.3 vs. 267.1 ± 15.4 ng/mL and 6.6 ± 0.7 vs. 10.3 ± 1.4 μg/mL respectively, p < 0.05) with similar results in sTNFR1, sVWF, sC5b-9 and C3a. Endothelial cells exposed to fetal sera from preeclampsia showed incremented expression of VCAM-1(38.1 ± 1.4% vs. 28.3 ± 1.6%, p < 0.01), ICAM-1 (12 ± 0.9% vs. 8.6 ± 0.6%, p < 0.05), VWF (43.5 ± 2.9% vs. 3.7 ± 0.3%, p < 0.05), and ROS (5 × 1013 ± 1 × 1012 vs. 3.5 × 1013 ± 1.4 × 1012, p < 0.01) with similar expression of VE-Cadherin and eNOS as compared to those exposed to control fetuses. While soluble C5b-9 was similar between the study groups (851.4 ± 177.5 vs. 751.4 ± 132.81 ng/mL, p > 0.05), significantly less C5b-9 deposits on endothelial cells were induced by fetal plasma from preeclamptic compared to normotensive mothers (fold change 0.08 ± 0.02 vs. 0.48 ± 0.13, p < 0.01). CONCLUSIONS High levels of endothelial adhesion molecules and oxidative stress products suggest endothelial damage and reduced in vitro deposition of C5b-9 indicates complement dysregulation in preeclampsia fetuses. More research is necessary to study the impact of preeclampsia on fetal vascular health and innate immunity.
Collapse
Affiliation(s)
- Alex Ramos
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de BarcelonaBarcelonaSpain
- Barcelona Endothelium TeamBarcelonaSpain
| | - Lina Youssef
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, IDIBAPSUniversity of BarcelonaBarcelonaSpain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona CampusBarcelonaSpain
| | - Patricia Molina
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de BarcelonaBarcelonaSpain
| | - Julia Martinez‐Sanchez
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de BarcelonaBarcelonaSpain
- Barcelona Endothelium TeamBarcelonaSpain
| | - Ana Belen Moreno‐Castaño
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de BarcelonaBarcelonaSpain
| | - Miquel Blasco
- Nephrology and Kidney Transplant Department, Center of Reference in Complex Glomerular Disease (CSUR), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de BarcelonaBarcelonaSpain
| | - Blanca De Moner
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de BarcelonaBarcelonaSpain
- Barcelona Endothelium TeamBarcelonaSpain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona CampusBarcelonaSpain
| | - Marta Tortajada
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, IDIBAPSUniversity of BarcelonaBarcelonaSpain
| | - Marta Camacho
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, IDIBAPSUniversity of BarcelonaBarcelonaSpain
| | - Maria Borrell
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, IDIBAPSUniversity of BarcelonaBarcelonaSpain
| | - Francesca Crovetto
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, IDIBAPSUniversity of BarcelonaBarcelonaSpain
| | - Marc Pino
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de BarcelonaBarcelonaSpain
| | - Gines Escolar
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de BarcelonaBarcelonaSpain
| | - Enric Carreras
- Barcelona Endothelium TeamBarcelonaSpain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona CampusBarcelonaSpain
| | - Eduard Gratacos
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, IDIBAPSUniversity of BarcelonaBarcelonaSpain
- Centre for Biomedical Research on Rare Diseases (CIBER‐ER)MadridSpain
| | - Maribel Diaz‐Ricart
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Universitat de BarcelonaBarcelonaSpain
- Barcelona Endothelium TeamBarcelonaSpain
| | - Marta Palomo
- Barcelona Endothelium TeamBarcelonaSpain
- Hematology External Quality Assessment Laboratory, Biomedical Diagnostic CenterHospital Clinic of BarcelonaBarcelonaSpain
| | - Fatima Crispi
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, IDIBAPSUniversity of BarcelonaBarcelonaSpain
- Centre for Biomedical Research on Rare Diseases (CIBER‐ER)MadridSpain
| |
Collapse
|
20
|
Soto LJ, Del Tufiño C, Macias-Pérez ME, Castro-García S, Jiménez-Cruz E, Bobadilla-Lugo RA. Epicatechin prevents preeclampsia-associated hypertension and oxidative stress. J Obstet Gynaecol Res 2025; 51:e16290. [PMID: 40355116 DOI: 10.1111/jog.16290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/28/2025] [Indexed: 05/14/2025]
Abstract
Preeclampsia (PE) is a frequent and dangerous multisystemic pregnancy complication, associated with blood pressure control. Some antioxidants, including chocolate-derived epicatechin, can effectively attenuate hypertensive disorders. AIM This study aimed to assess whether epicatechin or dark chocolate (DC) could revert vascular increased reactivity and oxidative stress, both features of an experimental PE model. METHODS Rats from healthy pregnant or PE groups received vehicle, epicatechin (10 mg/kg/day) po, or DC (1 g) po, administered on days 1-14 (early) or days 7-21(late) of pregnancy. Blood pressure was measured by the tail-cuff plethysmography method. Aorta contractility was evaluated using a conventional isolated organ bath, and oxidative stress was determined by nicotine adenine dinucleotide phosphate reduced (NADPH) serum activity. RESULTS Epicatechin and DC significantly reduced hypertension, decreased abdominal aorta contractility, and decreased NADPH activity of the PE animals. The effects were more evident when administered during the last 2 weeks of pregnancy. CONCLUSIONS Results suggest that epicatechin has a significant antihypertensive effect in PE mediated by an antioxidant activity that improves vascular contractility.
Collapse
Affiliation(s)
- Luis J Soto
- Escuela Superior de Medicina IPN, Mexico City, Mexico
| | | | | | | | | | | |
Collapse
|
21
|
Taylor BD, Haggerty CL, Amabebe E, Richardson LS. Current Evidence of Maternal Infection With Chlamydia trachomatis and Preeclampsia Risk. Am J Reprod Immunol 2025; 93:e70080. [PMID: 40298141 DOI: 10.1111/aji.70080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 01/13/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
Chlamydia trachomatis is the most common bacterial sexually transmitted infection (STI) in the United States. Ascending C. trachomatis can cause pelvic inflammatory disease (PID), potentially leading to subsequent infertility, ectopic pregnancy, and adverse pregnancy outcomes. There is growing evidence implicating infections (e.g., COVID-19, cytomegalovirus) in preeclampsia etiology, a maternal hypertensive disorder and leading cause of maternal morbidity and mortality. However, few studies have investigated the impact of STIs on preeclampsia risk. In this review, we provide an overview of the potential association between C. trachomatis and preeclampsia and identify future research needs through a critical evaluation of epidemiologic, in vitro, and in vivo studies. Unfortunately, current methodological limitations such as lower-quality study designs, selection bias, confounding bias, and variations in chlamydia diagnostic methods inhibit our understanding of the impact of C. trachomatis on preeclampsia. In addition, bench-side approaches such as animal models and in vitro studies have not elucidated the mechanisms linking C. trachomatis to preeclampsia. Understanding the biological pathways that could be disrupted by chlamydia is important as it may ultimately guide the development and use of novel therapeutics to augment standard antibiotic therapy to reduce pathology.
Collapse
Affiliation(s)
- Brandie DePaoli Taylor
- Department of Obstetrics and Gynecology, Division of Basic Science and Translational Research, University of Texas Medical Branch, Galveston, Texas, USA
- Academic Research, Advocate Aurora Research Institute, Milwaukee, Wisconsin, USA
| | - Catherine L Haggerty
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Emmanuel Amabebe
- Department of Obstetrics and Gynecology, Division of Basic Science and Translational Research, University of Texas Medical Branch, Galveston, Texas, USA
| | - Lauren S Richardson
- Department of Obstetrics and Gynecology, Division of Basic Science and Translational Research, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
22
|
Lazzari C, Bosco M, Garzon S, Simonetto C, Casarin J, Paolucci S, Cromi A, Ghezzi F, Uccella S. The impact of maternal age and body mass index on hypertensive disorders of pregnancy: Moving beyond the cut-off effect. Pregnancy Hypertens 2025; 40:101219. [PMID: 40311220 DOI: 10.1016/j.preghy.2025.101219] [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: 08/22/2024] [Revised: 03/07/2025] [Accepted: 04/22/2025] [Indexed: 05/03/2025]
Abstract
OBJECTIVES Maternal age (MA) and body mass index (BMI) are known risk factors for hypertensive disorders of pregnancy (HDP). Different threshold values are used to calculate preeclampsia risk scores, but the appropriateness of a cut point model has not been extensively evaluated. This is because the effects of both MA and BMI occur continuously. We aimed to investigate the relationship between MA, BMI, and HDP, respecting the continuous nature of the two independent variables. STUDY DESIGN We retrospectively selected all nulliparous women with singleton pregnancies who delivered after 22 gestational weeks from January 2005 to December 2019 (25,165 women). We used univariate and multivariable logistic regression analyses implementing linear, quadratic, cubic, and penalized splines functions to test, investigate, and describe the relationship between continuous BMI, continuous MA, and risk of HDP. MAIN OUTCOME MEASURES MA, BMI, and infertility treatments are independently associated with the risk of developing all HDP in nulliparous women. The impacts of MA and BMI on the risk of developing HDP occur gradually, and penalized splines functions resulted in the best model to describe these associations with a different model for each HDP. No interaction factors were retained between MA, BMI, and infertility treatments. CONCLUSIONS Given the relevance of HDP on maternal mortality, an accurate identification of women at increased risk of developing the disease is crucial. In an era where technology is rapidly advancing, new models for customized risk assessment of HDP, considering the continuous nature of MA and pregestational BMI, must be implemented.
Collapse
Affiliation(s)
- Cecilia Lazzari
- Unit of Obstetrics and Gynecology, Department of Surgery, Dentistry, Pediatrics, and Gynecology, AOUI Verona, University of Verona, Verona, Italy
| | - Mariachiara Bosco
- Unit of Obstetrics and Gynecology, Department of Surgery, Dentistry, Pediatrics, and Gynecology, AOUI Verona, University of Verona, Verona, Italy
| | - Simone Garzon
- Unit of Obstetrics and Gynecology, Department of Surgery, Dentistry, Pediatrics, and Gynecology, AOUI Verona, University of Verona, Verona, Italy.
| | - Chiara Simonetto
- Unit of Obstetrics and Gynecology, Department of Surgery, Dentistry, Pediatrics, and Gynecology, AOUI Verona, University of Verona, Verona, Italy
| | - Jvan Casarin
- Department of Obstetrics and Gynecology, "Filippo Del Ponte" Hospital, University of Insubria, Varese, Italy
| | - Sonia Paolucci
- Department of Obstetrics and Gynecology, "Filippo Del Ponte" Hospital, University of Insubria, Varese, Italy
| | - Antonella Cromi
- Department of Obstetrics and Gynecology, "Filippo Del Ponte" Hospital, University of Insubria, Varese, Italy
| | - Fabio Ghezzi
- Department of Obstetrics and Gynecology, "Filippo Del Ponte" Hospital, University of Insubria, Varese, Italy
| | - Stefano Uccella
- Unit of Obstetrics and Gynecology, Department of Surgery, Dentistry, Pediatrics, and Gynecology, AOUI Verona, University of Verona, Verona, Italy
| |
Collapse
|
23
|
Chen H, Li X, Xu X, Hu Y, Zheng J. Differentially expressed genes in the placentas with pre-eclampsia and fetal growth restriction using RNA sequencing and verification. J Perinat Med 2025:jpm-2025-0025. [PMID: 40285515 DOI: 10.1515/jpm-2025-0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025]
Abstract
OBJECTIVES Pre-eclampsia and fetal growth restriction (FGR) is a serious complication of pregnancy. Our study aimed to identify the DEGs and pathways in the placentas of patients with pre-eclampsia and FGR and investigated the relationships between these genes and clinical characteristics to elucidate the dysregulated placental mechanisms linked to pre-eclampsia and FGR. METHODS A total of 28 patients were enrolled in the study (6 individuals were selected for RNA sequencing and 22 participants was subjected to qRT-PCR). We used RNA sequencing to identify DEGs and pathways in the placenta. Ten DEGs were verified using qRT-PCR. The relationships between these DEGs and clinical characteristics were investigated using correlation analysis. RESULTS We detected significant molecular changes in the placentas of pre-eclampsia and FGR, encompassing diverse biological processes. SYDE1, HTRA1, and PAPPA2 expression was significantly upregulated, whereas MYL9, OLFML3, VTN, and AXNA8 expression was downregulated (p<0.05). The mRNA expression levels of SYDE1, HTRA1 and PAPPA2 displayed a positive correlation with systolic and diastolic blood pressure, LDH and LST, while exhibiting a negative correlation with fetal weight and serum albumin. And the mRNA expression levels of MYL9, OLFML3, VTN, and ANXA8 were significantly negatively correlated with systolic and diastolic blood pressure, LDH and LST, while exhibiting a positive correlation with fetal weight. CONCLUSIONS Our study found variations in gene expression and pathways in the placenta that may contribute to pre-eclampsia and FGR. Genes expressed in the placenta that are associated with clinical indicators could serve as potential biomarkers for assessing the occurrence of pre-eclampsia and FGR.
Collapse
Affiliation(s)
- Haiying Chen
- Department of Obstetrics and Gynecology, The Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, China
| | - Xiaoqing Li
- Department of Obstetrics and Gynecology, The Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, China
| | - Xiaoming Xu
- Department of Obstetrics and Gynecology, The Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, China
| | - Yanjun Hu
- Department of Obstetrics and Gynecology, The Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, China
| | - Jianqiong Zheng
- Department of Obstetrics and Gynecology, The Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, China
| |
Collapse
|
24
|
Barbouti A, Varvarousis DN, Kanavaros P. The Role of Oxidative Stress-Induced Senescence in the Pathogenesis of Preeclampsia. Antioxidants (Basel) 2025; 14:529. [PMID: 40427411 PMCID: PMC12108173 DOI: 10.3390/antiox14050529] [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: 03/07/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Preeclampsia is a hypertension condition of human pregnancy that poses a significant risk to pregnant women and their fetus. It complicates about 2-8% of human pregnancies worldwide and displays multifactorial pathogenesis, including increased placental oxidative stress because of disturbed utero-placental blood flow. Recent evidence suggests that increased oxidative stress promotes acceleration of the placental senescence which is implicated in the pathogenesis of preeclampsia. This review focuses on the mechanisms that lead to oxidative stress in preeclamptic patients and examines the role of oxidative stress-induced placental senescence in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Alexandra Barbouti
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (D.N.V.); (P.K.)
| | | | | |
Collapse
|
25
|
Liu L, Li X, Yang H, Xu F, Dong X. Bioinformatic Analysis of Apoptosis-Related Genes in Preeclampsia Using Public Transcriptomic and Single-Cell RNA Sequencing Datasets. J Inflamm Res 2025; 18:4785-4812. [PMID: 40224388 PMCID: PMC11992479 DOI: 10.2147/jir.s507660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
Purpose Apoptosis, which is crucial in preeclampsia (PE), affects trophoblast survival and placental function. We used transcriptomics and single-cell RNA sequencing (scRNA-seq) to explore apoptosis-related genes (ARGs) and their cellular mechanisms as potential PE biomarkers. Patients and Methods All the data included in this study were sourced from public databases. We used scRNA-seq and differential expression analysis, combined with five algorithms from the CytoHubba plugin, to identify ARGs as PE biomarkers. These were integrated into diagnostic nomograms. Mechanistic studies involved enrichment analysis and immune profiling. Biomarker expression was examined at the single-cell level, and verified in clinical samples by RT-qPCR. Results Cluster of Differentiation 44 (CD44), Macrophage migration inhibitory factor (MIF), PIK3R1, and Toll-like receptor 4 (TLR4) were identified as PE biomarkers. CD44 and TLR4 were down-regulated, while MIF and PIK3R1 were up-regulated. When integrated into the diagnostic nomogram, they showed clinical utility and affected cell functions. In the immune profile of PE, monocytes decreased, resting NK cells increased, and the activities of APC, checkpoint, T-cell co-stimulation, and MHC class I pathways reduced. ScRNA-seq identified 11 cell types, 10 of which were significantly different. Endothelial cell communication with other cell types decreased, while the interaction between common myeloid progenitors (CMP) and villous cytotrophoblasts (VCT) enhanced. The expression levels of CD44, MIF, and PIK3R1 in VCT were significantly different and key to PE. Their decrease in early PE and increase in late PE reflected the placenta's adaptation to adverse pregnancy conditions. Conclusion Four ARGs, CD44, MIF, PIK3R1, and TLR4, identified through comprehensive analyses, served as significant biomarkers for PE and offered insights into PE's cellular mechanisms of PE, providing valuable references for further research.
Collapse
Affiliation(s)
- Lingyan Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Department of Obstetrics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
| | - Xiuling Li
- Department of Obstetrics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
| | - Hongfen Yang
- Department of Obstetrics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
| | - Fei Xu
- Medical School, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Department of Pain Management, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
| | - Xudong Dong
- Department of Obstetrics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
| |
Collapse
|
26
|
Fei H, Lu X, Shi Z, Liu X, Yang C, Zhu X, Lin Y, Jiang Z, Wang J, Huang D, Liu L, Zhang S, Jiang L. Deciphering the preeclampsia-specific immune microenvironment and the role of pro-inflammatory macrophages at the maternal-fetal interface. eLife 2025; 13:RP100002. [PMID: 40152904 PMCID: PMC11952753 DOI: 10.7554/elife.100002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025] Open
Abstract
Preeclampsia (PE), a major cause of maternal and perinatal mortality with highly heterogeneous causes and symptoms, is usually complicated by gestational diabetes mellitus (GDM). However, a comprehensive understanding of the immune microenvironment in the placenta of PE and the differences between PE and GDM is still lacking. In this study, cytometry by time of flight indicated that the frequencies of memory-like Th17 cells (CD45RA-CCR7+IL-17A+CD4+), memory-like CD8+ T cells (CD38+CXCR3-CCR7+Helios-CD127-CD8+) and pro-inflam Macs (CD206-CD163-CD38midCD107alowCD86midHLA-DRmidCD14+) were increased, while the frequencies of anti-inflam Macs (CD206+CD163-CD86midCD33+HLA-DR+CD14+) and granulocyte myeloid-derived suppressor cells (gMDSCs, CD11b+CD15hiHLA-DRlow) were decreased in the placenta of PE compared with that of normal pregnancy (NP), but not in that of GDM or GDM&PE. The pro-inflam Macs were positively correlated with memory-like Th17 cells and memory-like CD8+ T cells but negatively correlated with gMDSCs. Single-cell RNA sequencing revealed that transferring the F4/80+CD206- pro-inflam Macs with a Folr2+Ccl7+Ccl8+C1qa+C1qb+C1qc+ phenotype from the uterus of PE mice to normal pregnant mice induced the production of memory-like IL-17a+Rora+Il1r1+TNF+Cxcr6+S100a4+CD44+ Th17 cells via IGF1-IGF1R, which contributed to the development and recurrence of PE. Pro-inflam Macs also induced the production of memory-like CD8+ T cells but inhibited the production of Ly6g+S100a8+S100a9+Retnlg+Wfdc21+ gMDSCs at the maternal-fetal interface, leading to PE-like symptoms in mice. In conclusion, this study revealed the PE-specific immune cell network, which was regulated by pro-inflam Macs, providing new ideas about the pathogenesis of PE.
Collapse
Affiliation(s)
- Haiyi Fei
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseHangzhouChina
- Zhejiang Key Laboratory of Precise Protection and Promotion of FertilityHangzhouChina
| | - Xiaowen Lu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseHangzhouChina
- Zhejiang Key Laboratory of Precise Protection and Promotion of FertilityHangzhouChina
| | - Zhan Shi
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseHangzhouChina
- Zhejiang Key Laboratory of Precise Protection and Promotion of FertilityHangzhouChina
| | - Xiu Liu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseHangzhouChina
- Zhejiang Key Laboratory of Precise Protection and Promotion of FertilityHangzhouChina
| | - Cuiyu Yang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseHangzhouChina
- Zhejiang Key Laboratory of Precise Protection and Promotion of FertilityHangzhouChina
| | - Xiaohong Zhu
- Department of Obstetrics and Gynecology, Zhejiang Xiaoshan HospitalHangzhouChina
| | - Yuhan Lin
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseHangzhouChina
- Zhejiang Key Laboratory of Precise Protection and Promotion of FertilityHangzhouChina
| | - Ziqun Jiang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseHangzhouChina
- Zhejiang Key Laboratory of Precise Protection and Promotion of FertilityHangzhouChina
| | - Jianmin Wang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseHangzhouChina
- Zhejiang Key Laboratory of Precise Protection and Promotion of FertilityHangzhouChina
| | - Dong Huang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseHangzhouChina
- Zhejiang Key Laboratory of Precise Protection and Promotion of FertilityHangzhouChina
| | - Liu Liu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseHangzhouChina
- Zhejiang Key Laboratory of Precise Protection and Promotion of FertilityHangzhouChina
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseHangzhouChina
- Zhejiang Key Laboratory of Precise Protection and Promotion of FertilityHangzhouChina
| | - Lingling Jiang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseHangzhouChina
- Zhejiang Key Laboratory of Precise Protection and Promotion of FertilityHangzhouChina
| |
Collapse
|
27
|
Paraskevas T, Gakis G, Papapanou M, Sergentanis TN, Sotiriadis A, Siristatidis CS. Statins for preventing preeclampsia. Cochrane Database Syst Rev 2025; 3:CD016133. [PMID: 40099754 PMCID: PMC11915783 DOI: 10.1002/14651858.cd016133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
OBJECTIVES This is a protocol for a Cochrane Review (intervention). The objectives are as follows: To evaluate the relative benefits and harms of statins for preeclampsia prevention in pregnant women.
Collapse
Affiliation(s)
| | - Georgios Gakis
- General University Hospital of Patras, University of Patras, Patras, Greece
| | - Michail Papapanou
- Second Department of Obstetrics and Gynecology, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodoros N Sergentanis
- Department of Public Health Policy, School of Public Health, University of West Attica, Athens, Greece
| | - Alexandros Sotiriadis
- Second Department of Obstetrics and Gynaecology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Charalampos S Siristatidis
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
28
|
Goularte PDS, Imperador CHL, Piedade FH, Guerin AN, Martins KDT, Boreski D, Gabriel EA, Gabriel SA, Tiezzi DG, Chin CM. Copper, Zinc, and Selenium Levels During Pregnancy and Their Relationship with Pre-Eclampsia Risk: A Systematic Review. Nutrients 2025; 17:1038. [PMID: 40292457 PMCID: PMC11946521 DOI: 10.3390/nu17061038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 04/30/2025] Open
Abstract
Pre-eclampsia (PE) affects about 5% of all pregnancies worldwide and is one of the leading causes of maternal and fetal morbidity and mortality. Some reports suggest that micronutrients may influence this condition, but there is no existing report analyzing data on copper (Cu), selenium (Se), and zinc (Zn) regarding quality and bias. Accurate information is crucial to support governmental healthcare actions, especially in developing countries such as Brazil. This study aims to investigate whether alterations in Zn, Cu, and Se levels in pregnant women contribute to the development of PE. We conducted the study following PRISMA guidelines and registered it on PROSPERO (CRD42022302298). We searched LILACS, PubMed/MEDLINE, EMBASE, and the Cochrane Library databases from January 2000 to January 2024. Of 1202 reports, 42 manuscripts were suitable for analysis (contained one or more micronutrients). A total of 76 individual analyses (by nutrient) were performed using Joanna Briggs Critical Appraisal and Cochrane Risk of Bias Tools. The analyses classified 69 reports as fair/low-quality with bias. Due to this, a meta-analysis was not conducted, as the results would not have accurately reflected reality. Of the high-quality reports, five on Cu showed conflicting results, while two studies on Zn found no differences in the level of this micronutrient between normal and PE pregnancies. No high-quality studies were identified for Se. The results highlight the need for robust guidelines for research involving micronutrients and PE to address this question effectively.
Collapse
Affiliation(s)
- Pietra dos Santos Goularte
- Advanced Research Center in Medicine (CEPAM), School of Medicine, Union of the Colleges of the Great Lakes (UNILAGO), Sao Jose do Rio Preto 15030-070, SP, Brazil; (P.d.S.G.); (C.H.L.I.); (F.H.P.); (A.N.G.); (K.d.T.M.); (E.A.G.); (S.A.G.); (D.G.T.)
| | - Carlos Henrique Lima Imperador
- Advanced Research Center in Medicine (CEPAM), School of Medicine, Union of the Colleges of the Great Lakes (UNILAGO), Sao Jose do Rio Preto 15030-070, SP, Brazil; (P.d.S.G.); (C.H.L.I.); (F.H.P.); (A.N.G.); (K.d.T.M.); (E.A.G.); (S.A.G.); (D.G.T.)
| | - Felipe Hassan Piedade
- Advanced Research Center in Medicine (CEPAM), School of Medicine, Union of the Colleges of the Great Lakes (UNILAGO), Sao Jose do Rio Preto 15030-070, SP, Brazil; (P.d.S.G.); (C.H.L.I.); (F.H.P.); (A.N.G.); (K.d.T.M.); (E.A.G.); (S.A.G.); (D.G.T.)
| | - Amanda Nascimento Guerin
- Advanced Research Center in Medicine (CEPAM), School of Medicine, Union of the Colleges of the Great Lakes (UNILAGO), Sao Jose do Rio Preto 15030-070, SP, Brazil; (P.d.S.G.); (C.H.L.I.); (F.H.P.); (A.N.G.); (K.d.T.M.); (E.A.G.); (S.A.G.); (D.G.T.)
| | - Karina de Toledo Martins
- Advanced Research Center in Medicine (CEPAM), School of Medicine, Union of the Colleges of the Great Lakes (UNILAGO), Sao Jose do Rio Preto 15030-070, SP, Brazil; (P.d.S.G.); (C.H.L.I.); (F.H.P.); (A.N.G.); (K.d.T.M.); (E.A.G.); (S.A.G.); (D.G.T.)
| | - Diogo Boreski
- Laboratory for Drug Design (LAPDESF), School of Pharmaceutical Sciences, University of São Paulo State (UNESP), Araraquara 14800-903, SP, Brazil;
| | - Edmo Atique Gabriel
- Advanced Research Center in Medicine (CEPAM), School of Medicine, Union of the Colleges of the Great Lakes (UNILAGO), Sao Jose do Rio Preto 15030-070, SP, Brazil; (P.d.S.G.); (C.H.L.I.); (F.H.P.); (A.N.G.); (K.d.T.M.); (E.A.G.); (S.A.G.); (D.G.T.)
| | - Sthefano Atique Gabriel
- Advanced Research Center in Medicine (CEPAM), School of Medicine, Union of the Colleges of the Great Lakes (UNILAGO), Sao Jose do Rio Preto 15030-070, SP, Brazil; (P.d.S.G.); (C.H.L.I.); (F.H.P.); (A.N.G.); (K.d.T.M.); (E.A.G.); (S.A.G.); (D.G.T.)
| | - Daniel Guimarães Tiezzi
- Advanced Research Center in Medicine (CEPAM), School of Medicine, Union of the Colleges of the Great Lakes (UNILAGO), Sao Jose do Rio Preto 15030-070, SP, Brazil; (P.d.S.G.); (C.H.L.I.); (F.H.P.); (A.N.G.); (K.d.T.M.); (E.A.G.); (S.A.G.); (D.G.T.)
- Gynecology Department, School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto 05508-220, SP, Brazil
| | - Chung Man Chin
- Advanced Research Center in Medicine (CEPAM), School of Medicine, Union of the Colleges of the Great Lakes (UNILAGO), Sao Jose do Rio Preto 15030-070, SP, Brazil; (P.d.S.G.); (C.H.L.I.); (F.H.P.); (A.N.G.); (K.d.T.M.); (E.A.G.); (S.A.G.); (D.G.T.)
- Laboratory for Drug Design (LAPDESF), School of Pharmaceutical Sciences, University of São Paulo State (UNESP), Araraquara 14800-903, SP, Brazil;
| |
Collapse
|
29
|
Ochoa JH, Cafici D. Fetal Doppler assessment in pregnancy. Best Pract Res Clin Obstet Gynaecol 2025; 100:102594. [PMID: 40187275 DOI: 10.1016/j.bpobgyn.2025.102594] [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: 08/10/2024] [Accepted: 02/24/2025] [Indexed: 04/07/2025]
Abstract
Fetal Doppler assessment plays a crucial role in monitoring the fetal well-being during pregnancy. This non-invasive technique assesses blood flow dynamics in key fetal vessels, namely the umbilical artery, middle cerebral artery, and ductus venosus. The umbilical artery Doppler provides valuable insights into placental function aiding in the early detection of fetal growth restriction and fetal distress. Assessment of the middle cerebral artery Doppler provides information on the adequacy of cerebral perfusion. It is highly sensitive to changes in fetal oxygenation and contributes to the management of advanced stages of early fetal growth restriction. It is also a valuable and sometimes standalone marker for late-term fetal hypoxic compromise and fetal anemia. Doppler evaluation of ductus venosus offers additional data for identifying cardiac compromise and predicting adverse perinatal outcomes. Incorporating these Doppler assessments into routine prenatal care enhances the ability to detect and manage fetal compromise, ultimately improving pregnancy outcomes.
Collapse
Affiliation(s)
- Jose H Ochoa
- Diagnus, Academy of Prenatal Diagnosis and Fetal Medicine, Córdoba, Argentina.
| | - Daniel Cafici
- SAUMB, Argentine Society of Ultrasound in Medicine and Biology, Argentina
| |
Collapse
|
30
|
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
|
31
|
Chen L, Cao Z, Qiao P, Liu X, Ying H. Impact of inter-twin growth discordance on preeclampsia: based on ultrasonic estimated fetal weight. Hypertens Res 2025; 48:894-903. [PMID: 39627393 PMCID: PMC11879843 DOI: 10.1038/s41440-024-02027-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/28/2024] [Accepted: 11/09/2024] [Indexed: 12/07/2024]
Abstract
A retrospective cohort study with 4396 twins who registered before 13 gestational weeks and delivered between January 2013 and December 2020 at Shanghai First Maternity and Infant Hospital, China, was conducted to clarify causal associations between inter-twin estimated fetal weight discordance and preeclampsia. Ultrasound measurements of fetal biometry were collected until the confirmation of preeclampsia diagnosis or the termination of pregnancy (when preeclampsia did not occur). Inter-twin discordance was divided into binary variables using cut-offs of 10%, 15%, and 20%. The associations between inter-twin discordance and preeclampsia were analyzed using generalized estimating equations and group-based trajectory modeling methods. The incidence of preeclampsia was 13.9%, among which 21.8% of cases were diagnosed at early onset and 55.3% at a severe stage. Inter-twin discordance based on estimated fetal weight during pregnancy was positively associated with preeclampsia. The associations were robust and constant by treating the discordance as continuous and binary. Two groups, the stable trajectory group, including 92% of participants, and the changing trajectory group, including 8% of participants, were divided according to the group-based trajectory models. Compared with the stable trajectory group, the risk of developing preeclampsia in the changing trajectory group increased by 50.3% (OR = 1.502, 95%CI: 1.073, 2.105). Subgroup analysis showed positive association primarily in early-onset preeclampsia (OR: 3.859, CI: 2.293, 6.494) and severe preeclampsia (OR: 1.896, CI: 1.264, 2.844) subgroups. These findings can provide a direction to reduce the incidence of preeclampsia in twin pregnancies, considering growth discordance as a high-risk factor in clinical practice.
Collapse
Affiliation(s)
- Lan Chen
- Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Obstetrics, Shidong Hospital affiliated with the University of Shanghai for Science and Technology, Shanghai, China
| | - Zhijuan Cao
- Department of Clinical Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ping Qiao
- Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaohua Liu
- Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Ying
- Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
32
|
Zhang C, Li J, Wang L, Yang P, Luo X. ALKBH1 knockdown promotes the growth, migration and invasion of HTR-8/SVneo cells through regulating the m5C modification PSMD14. Sci Rep 2025; 15:7345. [PMID: 40025166 PMCID: PMC11873043 DOI: 10.1038/s41598-025-91233-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/19/2025] [Indexed: 03/04/2025] Open
Abstract
Preeclampsia (PE) is a pregnancy disease characterized by insufficient invasion and growth of trophoblast cells. adeno-associated virus encoding alkB homolog 1 (ALKBH1) is a demethylase in 5-methylcytosine (m5C) methylation modification. This study was performed to explore the role of ALKBH1 in hypoxia treated human extravasated trophoblast cells. Hypoxia treated human extravasated trophoblast cells (HTR-8/SVneo) was used to simulate the occurrence of PE in vitro. The cells phenotype was detected by CCK-8 and Transwell assays. The m5c levels and m5C levels of PSMD14 were analyzed by m5C dot blot and M5C Me-RIP assays. Then, the interaction between ALKBH1 and PSMD14 were confirmed by RIP and dual-luciferase reporter assays. ALKBH1 was up-regulated in hypoxia treated HTR-8/SVneo cells. Additionally, ALKBH1 knockdown increased the m5C contents, cell viability, migration and invasion abilities of hypoxia treated HTR-8/SVneo cells. Furthermore, ALKBH1 knockdown increased the m5C and mRNA levels, and mRNA stability of PSMD14. RIP and dual-luciferase reporter assays demonstrated that ALKBH1 interacted with PSMD14. Besides, PSMD14 knockdown reversed the effects of ALKBH1 silencing on cell viability, migration and invasion abilities of hypoxia treated HTR-8/SVneo cells. ALKBH1 mediated m5C levels were decreased in the hypoxia treated HTR-8/SVneo cells, which further decreased the cell viability, migration and invasion abilities through targeting the PSMD14 levels.
Collapse
Affiliation(s)
- Caili Zhang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University or Maternal and Child Hospital of Henan Province, No.10, Kangfuqian Street, Zhengzhou City, 450001, Henan, China
| | - Jie Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University or Maternal and Child Hospital of Henan Province, No.10, Kangfuqian Street, Zhengzhou City, 450001, Henan, China
| | - Luwen Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University or Maternal and Child Hospital of Henan Province, No.10, Kangfuqian Street, Zhengzhou City, 450001, Henan, China
| | - Peifeng Yang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University or Maternal and Child Hospital of Henan Province, No.10, Kangfuqian Street, Zhengzhou City, 450001, Henan, China
| | - Xiaohua Luo
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University or Maternal and Child Hospital of Henan Province, No.10, Kangfuqian Street, Zhengzhou City, 450001, Henan, China.
| |
Collapse
|
33
|
Monteiro VNP, de Oliveira CA, Gomes Junior SC, do Cima LC, Naves WU, Diniz ALD, Araujo Júnior E, de Sá RAM. Ophthalmic Artery Doppler as a Predictor of Adverse Neonatal Outcomes in Women With Preeclampsia. JOURNAL OF CLINICAL ULTRASOUND : JCU 2025; 53:504-509. [PMID: 39588780 DOI: 10.1002/jcu.23899] [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: 06/30/2024] [Revised: 10/09/2024] [Accepted: 11/04/2024] [Indexed: 11/27/2024]
Abstract
OBJECTIVE This study aimed to evaluate the association between ophthalmic artery (OA) Doppler indices and adverse neonatal outcomes in women with pregnancies complicated by preeclampsia (PE). METHODS A prospective cross-sectional study involving 110 pregnant women who were diagnosed with PE in the first 24 h of hospitalization was conducted at three hospitals in southeastern Brazil from April 2020 to December 2022. The ophthalmic parameters analyzed were the resistance index (RI), pulsatility index (PI), first peak systolic velocity (PSV1), second peak systolic velocity (PSV2), ratio of PSV2 to PSV1 (PSV ratio or the peak ratio [PR]), and end-diastolic velocity (EDV). The outcome measures were neonatal death (during hospitalization), neonatal intensive care unit (ICU) admission, gestational age at delivery < 32 weeks, birth weight < 1500 g, birth weight < 10th percentile, the presence of comorbidities associated with prematurity, the use of invasive mechanical ventilation, an Apgar score at the 5th minute < 7.0, and signs of neurological impairment. RESULTS Pregnant women with severe features of PE showed higher values of systolic (152.8 ± 20.7 vs. 136.3 ± 11.9, p = 0.000) and diastolic (98.1 ± 12.8 vs. 88.1 ± 10.8, p = 0.000) blood pressure values and lower gestational age at admission (31.5 ± 3.8 vs. 33.7 ± 4.3, p = 0.009) than PE without severe features. No significant differences were found between the mean values of the OA Doppler indices when comparing the presence or absence of neonatal outcomes in the women with PE. CONCLUSIONS OA Doppler, while valuable for the prediction, diagnosis, and evaluation of adverse maternal outcomes, does not independently predict adverse neonatal outcomes in women with severe features of PE.
Collapse
Affiliation(s)
- Viviane Nascimento Pereira Monteiro
- Unit of Fetal Medicine, National Institute of Women's, Children's and Adolescents' Health Fernandes Figueira, Oswaldo Cruz Foundation (IFF/Fiocruz), Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Saint Clair Gomes Junior
- Unit of Fetal Medicine, National Institute of Women's, Children's and Adolescents' Health Fernandes Figueira, Oswaldo Cruz Foundation (IFF/Fiocruz), Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Wellington Ued Naves
- Department of Obstetrics and Gynecology, Federal University of Uberlândia (UFU), Uberlândia, Minas Gerais, Brazil
| | - Angélica Lemes Debs Diniz
- Department of Obstetrics and Gynecology, Federal University of Uberlândia (UFU), Uberlândia, Minas Gerais, Brazil
| | - Edward Araujo Júnior
- Department of Gynecology, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo, São Paulo, Brazil
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul, São Paulo, Brazil
| | - Renato Augusto Moreira de Sá
- Unit of Fetal Medicine, National Institute of Women's, Children's and Adolescents' Health Fernandes Figueira, Oswaldo Cruz Foundation (IFF/Fiocruz), Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Obstetrics, Fluminense Federal University (UFF), Niteroi, Rio de Janeiro, Brazil
| |
Collapse
|
34
|
Hurme V, Honkanen R, Backman K, Karvonen AM, Kirjavainen P, Keski-Nisula L. Earlier pregnancies in nulliparous women with current father and lower risks for preeclampsia and low-birth weight newborns. J Reprod Immunol 2025; 168:104431. [PMID: 39874628 DOI: 10.1016/j.jri.2025.104431] [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: 10/10/2024] [Revised: 12/18/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025]
Abstract
New paternity has been related to placenta-associated complications in pregnancy. We evaluated whether a lack of earlier pregnancies or deliveries with a current father are associated with the pregnancy, prenatal, and early neonatal outcomes after controlling for the most common maternal confounders in prospective birth cohort study. An online questionnaire was used to survey 4459 pregnant women from the Kuopio Birth Cohort in their third trimester. The topics included their history of paternity in current and earlier pregnancies. Data were combined with prenatal, perinatal, and early neonatal information. A multivariable logistic regression analysis was performed to compare the possible associations between selected pregnancy and early neonatal outcomes with respect to paternal change. Pregnant women with changed partners had higher rates of smoking during pregnancy and hypertension before pregnancy. In the adjusted analysis, primigravidas and nulliparous multigravidas with different father had the highest risks for preeclampsia (adjusted odds ratios (aORs) 4.46 and 2.69, respectively), low birth weight (aORs 3.15 and 2.25), and smallness for gestational age (aORs 2.23 and 2.16) compared to the parous controls. Nulliparous women who had earlier pregnancies with the current father had less preeclampsia or gestational hypertension, as well as low birth weight (aOR 0.42, 95 % confidence interval (CI) 0.21-0.82 and aOR 0.26, 95 % CI 0.08-0.87, respectively) compared to other nulliparas. Among parous women, most of the pregnancy, obstetric, and early neonatal outcomes were similar in the adjusted analysis, regardless of new changes in paternity.
Collapse
Affiliation(s)
- Veera Hurme
- Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Reetta Honkanen
- Department of Obstetrics and Gynaecology, North Karelia Central Hospital, Joensuu, Finland
| | - Katri Backman
- Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland; Department of Pediatrics, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Anne M Karvonen
- Department of Public Health, Finnish Institute for Health and Welfare, Kuopio, Finland
| | - Pirkka Kirjavainen
- Department of Public Health, Finnish Institute for Health and Welfare, Kuopio, Finland; Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Leea Keski-Nisula
- Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland; Department of Obstetrics and Gynaecology, Kuopio University Hospital, PL 100, Kuopio 70029, Finland.
| |
Collapse
|
35
|
Balhotra KS, Sibai BM. Aspirin dosage for preeclampsia prophylaxis: an argument for 81-mg dosing. Am J Obstet Gynecol MFM 2025; 7:101568. [PMID: 39586473 DOI: 10.1016/j.ajogmf.2024.101568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 11/27/2024]
Abstract
Research conducted over the past few decades has shown that low-dose aspirin can effectively reduce the risk of developing preeclampsia. Consequently, numerous prominent organizations have adopted the recommendation to use low-dose aspirin during pregnancy to prevent preeclampsia. However, the optimal dosage of low-dose aspirin (81mg versus 162mg) remains a subject of debate. Currently, there is insufficient high-quality data to justify the use of a higher dosage of low-dose aspirin. In this review, we review the existing evidence that supports the continued use of 81mg of aspirin over a higher dose and emphasize the need for high-quality research to alter current recommendations.
Collapse
Affiliation(s)
- Kimen S Balhotra
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Texas Health Science Center at Houston, Houston, TX.
| | - Baha M Sibai
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Texas Health Science Center at Houston, Houston, TX
| |
Collapse
|
36
|
Mills AN, Dubois B, Lesseur C, Rommel AS, Tubassum R, Kaplowitz E, Boychuk N, Stern T, Chen J, Lieb W, Janevic T, Jessel RH. Impact of antenatal SARS-CoV-2 infection on development of hypertensive disorders of pregnancy in a large, diverse, cohort. Pregnancy Hypertens 2025; 39:101205. [PMID: 40010184 DOI: 10.1016/j.preghy.2025.101205] [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: 01/19/2024] [Revised: 02/09/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025]
Abstract
OBJECTIVES Various studies have noted an association between antenatal SARS-CoV-2 infection and increased risk for development of hypertensive disorders of pregnancy (HDP). Both disease processes have been shown to involve endothelial dysfunction systemically and in the placenta, suggesting common pathogenesis. We aim to further investigate this association in a diverse urban population. STUDY DESIGN Generation C is a prospective pregnancy cohort study at a large academic institution in NYC established between April 2020 and February 2022. SARS-CoV-2 infection during pregnancy was ascertained using a combination of spike and nucleocapsid IgG antibodies, RT-PCR testing, and electronic medical record (EMR) diagnoses. Maternal demographic and medical data were ascertained from the EMR and/or self-report survey. MAIN OUTCOME MEASURES The primary outcome was HDP defined using the American College of Obstetrics and Gynecology diagnostic criteria. Covariates included maternal age ≥ 35 years, BMI ≥ 30, high social vulnerability index based on patient zip code, maternal chronic hypertension, pregestational diabetes, and nulliparity. Univariable and multivariable logistic regression was used to examine the association between antenatal SARS-CoV-2 infection and HDP. RESULTS Among the 2402 participants, 15.4 % (369) were infected with SARS-CoV-2 during pregnancy and 18.2 % (67/369) of those exposed developed an HDP. In participants without evidence of antenatal SARS-COV-2 infection, 18.0 % (365/2033) developed an HDP. In an adjusted multivariable model, antenatal SARS-CoV-2 infection was not associated with HDP (aOR 0.89; 95 % CI, 0.65-1.22). CONCLUSIONS This study did not find an increased risk of HDP associated with antenatal SARS-CoV-2 infection in a diverse prospective cohort.
Collapse
Affiliation(s)
- Alexandra N Mills
- Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Bethany Dubois
- Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York City, NY, USA.
| | - Corina Lesseur
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Anna-Sophie Rommel
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Rushna Tubassum
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Elianna Kaplowitz
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Natalie Boychuk
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York City, NY, USA
| | - Toni Stern
- Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Jia Chen
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Whitney Lieb
- Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York City, NY, USA; Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Teresa Janevic
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York City, NY, USA
| | - Rebecca H Jessel
- Division of Maternal Fetal Medicine, Department of Obstetrics & Gynecology, New York University Langone Health, New York, New York, USA
| |
Collapse
|
37
|
Wu X, Hong J, Hong L. The Deubiquitinating Enzyme USP4 Promotes Trophoblast Dysfunction by Stabilizing RYBP. Cell Biochem Biophys 2025; 83:929-939. [PMID: 39405024 DOI: 10.1007/s12013-024-01525-0] [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] [Accepted: 09/07/2024] [Indexed: 03/03/2025]
Abstract
Previous studies have suggested that impaired spiral artery remodeling, placental dysfunction, and insufficient trophoblast infiltration are the etiology and pathogenesis of Preeclampsia (PE). Ring 1 and YY1 binding protein (RYBP) has been reported to be associated with trophoblast dysfunction. However, the molecular mechanism of RYBP involved in trophoblasts in the pathogenesis of PE is poorly defined. RYBP and Ubiquitin-specific peptidase 4 (USP4) mRNA levels were determined using real-time quantitative polymerase chain reaction (RT-qPCR). RYBP, USP4, p-PI3K, PI3K, p-AKT, and AKT protein levels were measured using western blot assay. Cell viability, proliferation, apoptosis, invasion, and migration were assessed using 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT), 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, transwell, and wound healing assays. After ubibrowser database analysis, the interaction between USP4 and RYBP was verified using Co-immunoprecipitation (CoIP) assay. RYBP and USP4 expression were upregulated in placental tissues from PE patients. By using JEG-3 and HTR-8/SVneo trophoblast cells, RYBP overexpression or USP4 upregulation could hinder cell viability, proliferation, invasion, migration, and promote apoptosis. Mechanistically, USP4 could trigger the deubiquitination of RYBP and prevent its degradation. In addition, USP4 repressed the PI3K/AKT signaling pathway by regulating RYBP. In total, Decreased USP4-mediated ubiquitination results in an adverse impact on trophoblast function by enhancing RYBP expression, providing a novel therapeutic target for PE.
Collapse
Affiliation(s)
- Xuandi Wu
- Department of Obstetrics, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, China
| | - Jia Hong
- Department of Obstetrics, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China.
| | - Liang Hong
- Department of Emergency, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
38
|
Nobrega GM, Pietro L, Dariva SL, Vasconcelos-Silva IA, Manari MP, Polli B, Simões AB, de Almeida JS, Moschetta R, Ribeiro-do-Valle CC, Siqueira Guida JP, Souza RT, Cecatti JG, Mysorekar IU, Picoloto AS, Costa ML. Preeclampsia biomarkers (sFlt-1/PlGF) dynamics are not disrupted by SARS-CoV-2 infection during pregnancy in a hypertensive disorder SARS-CoV-2 vaccinated cohort. Pregnancy Hypertens 2025; 39:101196. [PMID: 39892161 DOI: 10.1016/j.preghy.2025.101196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 12/05/2024] [Accepted: 01/23/2025] [Indexed: 02/03/2025]
Abstract
OBJECTIVES To analyze maternal and perinatal outcomes and serum levels of the preeclampsia (PE) biomarkers sFlt-1 and PlGF in pregnant women with hypertensive disorders vaccinated against SARS-CoV-2, with or without confirmed COVID-19 during pregnancy. METHODS Multicenter (two-center) prospective cohort study secondary analysis. The cohort comprised pregnant women with hypertensive disorders who received SARS-CoV-2 vaccination, assessed from August 2021 to December 2022. Key variables included sociodemographic information, clinical background, maternal and perinatal outcomes, and biomarkers serum concentrations. A sFlt-1/PlGF ratio ≥ 38 was the threshold for predicting PE. The study compared outcomes based on the timing of SARS-CoV-2 infection and PE clinical onset. RESULTS For biomarker analysis, 170 women provided serum samples: 31 had a confirmed COVID-19 during pregnancy, while 139 did not. Among these 170 women, 86 had chronic hypertension, and 100 developed PE. There were no significant differences in sociodemographic characteristics and gestational outcomes between the groups. The dynamics of the sFlt-1/PlGF ratio were similar regardless of COVID-19. Cesarean delivery was the most common delivery method in both groups, and there was a high rate of preterm births. No neonatal or maternal deaths were recorded. CONCLUSIONS The study suggests that pregnant women with hypertensive disorders who have been vaccinated against SARS-CoV-2 and subsequently infected show comparable maternal and perinatal outcomes and PE biomarker levels to those who were not infected. These results suggest that SARS-CoV-2 vaccination is protective for pregnant women, potentially reducing the association with a PE-like syndrome in severe cases of COVID-19 among those who are unvaccinated.
Collapse
Affiliation(s)
- Guilherme Moraes Nobrega
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas Campinas SP Brazil; Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Luciana Pietro
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas Campinas SP Brazil; Institute of Health Sciences, Paulista University, Campinas, SP, Brazil
| | - Sarah Luiza Dariva
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas Campinas SP Brazil
| | | | - Monique Possari Manari
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas Campinas SP Brazil
| | - Barbara Polli
- Department of Obstetrics and Gynecology, School of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Arthur Becker Simões
- Department of Obstetrics and Gynecology, School of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Julia Stucker de Almeida
- Department of Obstetrics and Gynecology, School of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Roberta Moschetta
- Department of Obstetrics and Gynecology, School of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Jose Paulo Siqueira Guida
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas Campinas SP Brazil
| | - Renato Teixeira Souza
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas Campinas SP Brazil
| | - Jose Guilherme Cecatti
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas Campinas SP Brazil
| | - Indira U Mysorekar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ana Selma Picoloto
- Department of Obstetrics and Gynecology, School of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maria Laura Costa
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas Campinas SP Brazil.
| |
Collapse
|
39
|
Ku YL, Chen YY, Yang YH, Lee CP, Chen KJ, Ou YC. Would internal iliac artery occlusion cause gestational hypertension in the following pregnancy? A population-based study from 2008 to 2017. Taiwan J Obstet Gynecol 2025; 64:303-312. [PMID: 40049816 DOI: 10.1016/j.tjog.2024.12.009] [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] [Accepted: 12/20/2024] [Indexed: 05/13/2025] Open
Abstract
OBJECTIVE In animal models, internal iliac artery occlusion caused gestational hypertension; however, whether this phenomena occurs in humans is still unknown. MATERIALS AND METHODS This retrospective cohort study used data from the Birth Certificate Application of Taiwan and linked to the National Health Insurance Research Database and Taiwan Maternal and Child Health Database from 2008 to 2017. Women who underwent internal iliac artery occlusion before pregnant were identified according to diagnosis and procedure codes. The occlusion group included 328 births in 286 women with a history of internal iliac artery occlusion, and the non-occlusion control group included 2,024,882 births in 1,391,288 women. RESULTS There were no significant differences in gestational hypertension-associated diseases including preeclampsia, eclampsia and HELLP syndrome between the occlusion and non-occlusion groups (4.3 % vs 3.4 %, p = 0.4). The adjusted odds ratios (ORs) of placental previa, placenta accreta spectrum and stillbirth were 1.69 (95 % confidence interval [CI] = 1.12-2.56), 3.99 (95 % CI = 2.52-6.31), 2.57 (95 % CI = 1.13-5.83), respectively, with the non-occlusion group as reference. The adjusted ORs of preterm delivery in the occlusion group were 1.48 (95 % CI = 1.08-2.04) and 2.79 (95 % CI = 1.62-4.82) for a gestational age below 37 weeks and 32 weeks, respectively. CONCLUSION Women who underwent internal iliac artery occlusion did not have a higher risk of gestational hypertension and related disease. Their offspring also had similar risks of small for gestational age, poor Apgar score, birth defects and neonatal mortality within 28 days. However, their risks of placental previa, placenta accreta spectrum, stillbirth and preterm delivery were increased.
Collapse
Affiliation(s)
- Yu-Lun Ku
- Department of Gynecology and Obstetrics, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan; Department of Early Childhood Care and Education, Shu-Zen Junior College of Medicine and Management, Kaohsiung City, Taiwan
| | - Ying-Yi Chen
- Department Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yao-Hsu Yang
- Health Information and Epidemiology Laboratory, Chang Gung Memorial Hospital, Chiayi, Taiwan; School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Traditional Chinese Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Chuan-Pin Lee
- Health Information and Epidemiology Laboratory, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Ko-Jung Chen
- Health Information and Epidemiology Laboratory, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yu-Che Ou
- Department of Gynecology and Obstetrics, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan; Department Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
40
|
Ma G, Chen Z, Xie Z, Liu J, Xiao X. Mechanisms underlying changes in intestinal permeability during pregnancy and their implications for maternal and infant health. J Reprod Immunol 2025; 168:104423. [PMID: 39793281 DOI: 10.1016/j.jri.2025.104423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 12/01/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Proper regulation of intestinal permeability is essential for maintaining the integrity of the intestinal mucosal barrier. An abnormal increase in permeability can significantly contribute to the onset and progression of various diseases, including autoimmune disorders, metabolic conditions, allergies, and inflammatory bowel diseases. The potential connection between intestinal permeability and maternal health during pregnancy is increasingly recognized, yet a comprehensive review remains lacking. Pregnancy triggers a series of physiological structural adaptations and significant hormonal fluctuations that collectively contribute to an increase in intestinal permeability. Although an increase in intestinal permeability is typically a normal physiological response during pregnancy, an abnormal rise is associated with immune dysregulation, metabolic disorders, and various pregnancy-related complications, such as recurrent pregnancy loss, gestational diabetes mellitus, overweight and obesity during pregnancy, intrahepatic cholestasis of pregnancy, and preeclampsia. This paper discusses the components of the intestinal mucosal barrier, the concept of intestinal permeability and its measurement methods, and the mechanisms and physiological significance of increased intestinal permeability during pregnancy. It thoroughly explores the association between abnormal intestinal permeability during pregnancy and maternal diseases, aiming to provide evidence for the pathophysiology of disease development in pregnant women. Additionally, the paper examines intervention methods, such as gut microbiota modulation and nutritional interventions, to regulate intestinal permeability during pregnancy, improve immune and metabolic states, and offer feasible strategies for the prevention and adjuvant treatment of clinical pregnancy complications.
Collapse
Affiliation(s)
- Guangyu Ma
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Zhongsheng Chen
- Department of Colorectal Cancer Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Zhuojun Xie
- General Medicine Department, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - JinXiang Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Xiaomin Xiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China.
| |
Collapse
|
41
|
Nobakht N, Afshar Y, Vaseghi M, Li Z, Donangelo I, Lavretsky H, Mok T, Han CS, Nicholas SB. Hypertension Management in Women With a Multidisciplinary Approach. Mayo Clin Proc 2025; 100:514-533. [PMID: 39736047 PMCID: PMC12013344 DOI: 10.1016/j.mayocp.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 08/25/2024] [Accepted: 10/11/2024] [Indexed: 12/31/2024]
Abstract
Current clinical practice guidelines were established by several organizations to guide the diagnosis and treatment of hypertension in men and women in a similar manner despite data demonstrating differences in underlying mechanisms. Few publications have provided a contemporary and comprehensive review focused on characteristics of hypertension that are unique to women across their life spectrum. We performed a computerized search using PubMed, OVID, EMBASE, and Cochrane library databases between 1995 and 2023 that highlighted relevant clinical studies, challenges to the management of hypertension in women, and multidisciplinary approaches to hypertension control in women, including issues unique to racial and ethnic minority groups. Despite our current understanding of underlying mechanisms and strategies to manage hypertension in women, numerous challenges remain. Here, we discuss potential factors contributing to hypertension in women, differences related to effects of lifestyle modifications and drug therapy between men and women, the impact of sleep, and the importance of recognizing disparities in socioeconomic conditions and access to care. This review outlines several opportunities for future studies to fill gaps in knowledge to achieve optimal control of hypertension in women using a multidisciplinary approach, particularly related to sex-specific treatment approaches while considering socioeconomic conditions and life stages from premenopause through the transition to menopause.
Collapse
Affiliation(s)
- Niloofar Nobakht
- Division of Nephrology, Department of Medicine, at the David Geffen School of Medicine at University of California, Los Angeles, CA, USA.
| | - Yalda Afshar
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, at the David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Marmar Vaseghi
- Division of Cardiology, Department of Medicine, at the David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Zhaoping Li
- Division of Clinical Nutrition, Department of Medicine, at the David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Ines Donangelo
- Division of Endocrinology, Department of Medicine, at the David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Helen Lavretsky
- Department of Psychiatry at the David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Thalia Mok
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, at the David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Christina S Han
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, at the David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Susanne B Nicholas
- Division of Nephrology, Department of Medicine, at the David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| |
Collapse
|
42
|
Afrose D, Johansen MD, Nikolic V, Karadzov Orlic N, Mikovic Z, Stefanovic M, Cakic Z, Hansbro PM, McClements L. Evaluating oxidative stress targeting treatments in in vitro models of placental stress relevant to preeclampsia. Front Cell Dev Biol 2025; 13:1539496. [PMID: 40109359 PMCID: PMC11920713 DOI: 10.3389/fcell.2025.1539496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 01/20/2025] [Indexed: 03/22/2025] Open
Abstract
Background Preeclampsia is a complex pregnancy disorder characterized by the new onset of hypertension and organ dysfunction, often leading to significant maternal and fetal morbidity and mortality. Placental dysfunction is a hallmark feature of preeclampsia, which is often caused by inappropriate trophoblast cell function in association with oxidative stress, inflammation and/or pathological hypoxia. This study explores the role of oxidative stress in trophoblast cell-based models mimicking the preeclamptic placenta and evaluates potential therapeutic strategies targeting these mechanisms. Methods Uric acid (UA) and malondialdehyde (MDA) concentrations were measured in human plasma from women with preeclampsia (n = 24) or normotensive controls (n = 14) using colorimetric assays. Custom-made first trimester trophoblast cell line, ACH-3P, was exposed to various preeclampsia-like stimuli including hypoxia mimetic (dimethyloxalylglycine or DMOG, 1 mM), inflammation (tumour necrosis factor or TNF-α, 10 ng/mL) or mitochondria dysfunction agent, (Rhodamine-6G or Rho-6G, 1 μg/mL), ± aspirin (0.5 mM), metformin (0.5 mM), AD-01 (100 nM) or resveratrol (15 µM), for 48 h. Following treatments, UA/MDA, proliferation (MTT), wound scratch and cytometric bead, assays, were performed. Results Overall, MDA plasma concentration was increased in the preeclampsia group compared to healthy controls (p < 0.001) whereas UA showed a trend towards an increase (p = 0.06); when adjusted for differences in gestational age at blood sampling, MDA remained (p < 0.001) whereas UA became (p = 0.03) significantly correlated with preeclampsia. Our 2D first trimester trophoblast cell-based in vitro model of placental stress as observed in preeclampsia, mimicked the increase in UA concentration following treatment with DMOG (p < 0.0001), TNF-α (p < 0.05) or Rho-6G (p < 0.001) whereas MDA cell concentration increased only in the presence of DMOG (p < 0.0001) or Rho-6G (p < 0.001). Metformin was able to abrogate DMOG- (p < 0.01), Rho-6G- (p < 0.0001) or TNF-α- (p < 0.01) induced increase in UA, or DMOG- (p < 0.0001) or TNF-α- (p < 0.05)induced increase in MDA. AD-01 abrogated UA or MDA increase in the presence of TNF-α (p < 0.001) or Rho-6G (p < 0.001)/DMOG (p < 0.0001), respectively. The preeclampsia-like stimuli also mimicked adverse impact on trophoblast cell proliferation, migration and inflammation, most of which were restored with either aspirin, metformin, resveratrol, or AD-01 (p < 0.05). Conclusion Our 2D in vitro models recapitulate the response of the first trimester trophoblast cells to preeclampsia-like stresses, modelling inappropriate placental development, and demonstrate therapeutic potential of repurposed treatments.
Collapse
Affiliation(s)
- Dinara Afrose
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Matt D Johansen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Valentina Nikolic
- Department of Pharmacology with Toxicology, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Natasa Karadzov Orlic
- Department of Gynaecology and Obstetrics, Narodni Front, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Zeljko Mikovic
- Department of Gynaecology and Obstetrics, Narodni Front, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milan Stefanovic
- Department of Gynaecology and Obstetrics, Clinical Centre Nis, Nis, Serbia
- Department of Gynaecology and Obstetrics, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Zoran Cakic
- Department of Gynaecology and Obstetrics, General Hospital of Leskovac, Leskovac, Serbia
| | - Philip M Hansbro
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
43
|
Ruan F, Wang Y, Ying X, Liu Y, Xu J, Zhao H, Zhu Y, Wen P, Li X, Zhou Q, Huang H. Bioinformatics analysis of shared biomarkers and immune pathways of preeclampsia and periodontitis. BMC Pregnancy Childbirth 2025; 25:217. [PMID: 40016711 PMCID: PMC11866586 DOI: 10.1186/s12884-025-07277-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 02/03/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Epidemiological evidence indicates that preeclampsia (PE) is associated with comorbidities such as periodontitis (PD). However, the underlying mechanism remains unclear. To enhance our understanding of their co-pathogenesis, this research investigated the shared biomarkers and pathological mechanisms. METHODS We systematically retrieved transcriptomic datasets from the Gene Expression Omnibus database. These datasets encompass a comparative analysis of the periodontium with and without PD and of the placenta with and without PE. Differentially Expressed Genes Analysis and Weighted Gene Go-expression Network Analysis (WGCNA) were used to identify the key crosstalk genes in patients with PD and PE. The functional characterisation of these genes was performed using enrichment analysis. Protein-protein interaction networks and machine learning methods were leveraged to identify shared hub genes. The XG-Boost algorithm was applied to construct diagnostic models to gain insight into disease aetiology. The identified genes were validated by single-cell RNA sequencing to ensure their robustness and biological relevance. RESULTS A total of 55 key crosstalk genes were identified, which were primarily enriched in immune-related pathways by using limma and WGCNA. Among them, twenty-four shared hub genes were identified using protein-protein interaction analysis and machine learning methods. The diagnostic model constructed using immune-related genes outperformed the other two models (area under the receiver operating characteristic curve [ROC] = 0.7786 and 0.7454 for PE and PD, respectively). Pathways involving these genes were mapped using the Kyoto Encyclopedia of Genes and Genomes analysis. In addition, single-cell RNA sequencing analysis showed that the expression of BIN2, LYN, PIK3AP1, and NEDD9 in neutrophils was significantly downregulated, and LYN in fibroblasts and endothelial cells was consistently upregulated. CONCLUSIONS Shared hub genes and immunologic pathways were identified in PE and PD, characterised by crosstalk between BIN2, LYN, NEDD9, and PIK3AP1, suggesting the pathogenesis of PE and PD, which could pave the way for the development of effective diagnostic, treatment, and management strategies.
Collapse
Affiliation(s)
- Fangyi Ruan
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yinan Wang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xiang Ying
- Department of Prenatal Diagnostic Center, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yadan Liu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jinghui Xu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Huanqiang Zhao
- Institute of Maternal and Child Medicine, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong, China
- Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Yawei Zhu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Ping Wen
- Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China.
| | - Xiaotian Li
- Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China.
| | - Qiongjie Zhou
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
44
|
Freedman AA, Miller GE, Franklin AD, Keenan-Devlin LS, Gilman SE, Borders A, Khan SS, Ernst LM. Placental Pathology and Blood Pressure at Age 7: A Longitudinal Discordant Twin Analysis. Arterioscler Thromb Vasc Biol 2025; 45:312-322. [PMID: 39697173 PMCID: PMC11771519 DOI: 10.1161/atvbaha.124.321666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Evidence suggests that the intrauterine environment shapes offspring cardiovascular disease risk. Although placental dysfunction may be an important pathophysiologic pathway, numerous parental and pregnancy characteristics that influence offspring blood pressure are strong confounders of the mechanistic role of the placenta in observational analyses of singletons. Therefore, we leverage twin- and sibling-based comparison designs to determine whether placental pathology is associated with offspring blood pressure at age 7 while mitigating major sources of confounding. METHODS Data are from pregnant participants and their offspring in the Collaborative Perinatal Project, a longitudinal pregnancy cohort conducted from 1959 to 1965 in the United States. After delivery, placentas were systematically examined for lesions indicative of maternal vascular malperfusion (MVM) and acute inflammation. Blood pressure was assessed at a follow-up research visit when the offspring were 7 years old. Linear fixed-effects models were used to estimate associations between within-twin or sibling discordance in placental pathology and differences in blood pressure at age 7. RESULTS Overall, 193 twin pairs were eligible for inclusion, and 23.3% had placentas discordant for MVM. In a fixed-effect analysis, a twin with high-grade MVM had a higher systolic blood pressure Z score by 0.56 SDs than their co-twin without MVM (95% CI, 0.06-1.05) or a 5.7-mm Hg difference (95% CI, 0.6-10.8). Associations were consistent in a sensitivity analysis restricted to dichorionic twins and in a secondary analysis of 759 MVM-discordant sibling pairs. Acute placental inflammation was not associated with blood pressure at age 7. CONCLUSIONS MVM in the placenta is associated with higher offspring blood pressure in mid-childhood, independent of parental and pregnancy characteristics that twins have in common. The findings support the role of the placenta and the intrauterine environment in the developmental origins of cardiovascular health.
Collapse
Affiliation(s)
- Alexa A Freedman
- Department of Obstetrics and Gynecology, Endeavor Health, Evanston, IL (A.A.F.)
- Now with Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (A.A.F.)
| | - Gregory E Miller
- Institute for Policy Research (G.E.M.), Northwestern University, Evanston, IL
- Department of Psychology (G.E.M.), Northwestern University, Evanston, IL
| | - Andrew D Franklin
- Department of Pediatrics, Endeavor Health, Evanston, IL (A.D.F.), Endeavor Health, Evanston, IL
| | - Lauren S Keenan-Devlin
- Department of Obstetrics and Gynecology (L.S.K.-D., A.B.), Endeavor Health, Evanston, IL
- Department of Obstetrics and Gynecology, The University of Chicago Pritzker School of Medicine, Chicago, IL (L.S.K.-D., A.B.)
| | - Stephen E Gilman
- Social and Behavioral Sciences Branch, Division of Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (S.E.G.)
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (S.E.G.)
| | - Ann Borders
- Department of Obstetrics and Gynecology (L.S.K.-D., A.B.), Endeavor Health, Evanston, IL
- Department of Obstetrics and Gynecology, The University of Chicago Pritzker School of Medicine, Chicago, IL (L.S.K.-D., A.B.)
- Department of Medical Social Sciences (A.B.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Sadiya S Khan
- Department of Preventive Medicine (S.S.K.), Northwestern University Feinberg School of Medicine, Chicago, IL
- Division of Cardiology, Department of Medicine (S.S.K.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Linda M Ernst
- Department of Pathology and Laboratory Medicine (L.M.E.), Endeavor Health, Evanston, IL
| |
Collapse
|
45
|
Chen Y, Gao Z, Wang L, Duan R, Hao H, Jia R, Ma H, Gao R, Su M, Yang H, Tu Z. The effects of metformin on inflammation and apoptosis in rats with preeclampsia. J Hypertens 2025; 43:255-263. [PMID: 39351873 DOI: 10.1097/hjh.0000000000003892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/18/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Defined clinically by elevated blood pressure along with either proteinuria and/or maternal organ dysfunction, representing a major cause of morbidity and mortality pregnant women and newborns. Metformin (MET), an oral antidiabetic medication, has been shown to prevent preeclampsia (PE) through various mechanisms, including reducing inflammation, improving endothelial dysfunction, improving mitochondrial function, and altering cellular homeostasis and energy metabolism. Herein, we explored the role of MET in PE and its underlying molecular mechanisms using in in vivo experiments. METHODS RT-qPCR, Western blot (WB), and immunohistochemistry (IHC) were conducted to assess the mRNA or protein expression of genes related to mitochondrial apoptosis. Additionally, ELISA was conducted to quantify the expression of mitochondrial apoptosis and inflammation-related genes, as well as PE biomarkers. RESULTS Treatment with MET in PE rats ameliorated hypertension and proteinuria, altered the expression of PE biomarkers, and significantly inhibited L-NAME-induced inflammation and cell apoptosis. MET modulated the levels of inflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin (IL)-6, and IL-10, mitigating inflammation in PE rats. Furthermore, MET regulated mitochondrial outer membrane permeability (MOMP), thereby reducing cell apoptosis occurring in the mitochondrial pathway of PE rats. CONCLUSIONS This study demonstrates that MET alleviates inflammation and cell apoptosis in PE rats by modulating the expression of inflammatory factors and MOMP. Our results indicate that MET has huge therapeutic potential against PE.
Collapse
Affiliation(s)
| | | | - Liyuan Wang
- First Hospital of Shanxi Medical University, Taiyuan, China
| | - Ruiyun Duan
- First Hospital of Shanxi Medical University, Taiyuan, China
| | | | | | - Huijing Ma
- First Hospital of Shanxi Medical University, Taiyuan, China
| | - Ruifan Gao
- First Hospital of Shanxi Medical University, Taiyuan, China
| | - Min Su
- First Hospital of Shanxi Medical University, Taiyuan, China
| | - Hailan Yang
- First Hospital of Shanxi Medical University, Taiyuan, China
| | - Zengrong Tu
- First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
46
|
Winther M, Dziegiel MH, Thorsen SU. Preeclampsia and fetal growth restriction: does novel proteomics reveal immunological possible candidate biomarkers? Curr Opin Lipidol 2025; 36:21-26. [PMID: 39607830 DOI: 10.1097/mol.0000000000000965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
PURPOSE OF REVIEW The aim of this review is to explore a possible link between immunological candidate proteins, identified through modern proteomic techniques, and preeclampsia (PE) and fetal growth restriction (FGR). RECENT FINDINGS Proteomics has become a promising tool in the search for disease pathways, drug targets, and biomarkers. PE and FGR are adverse pregnancy complications with supposed immunological involvement in their pathogenesis, but no circulating immunological biomarkers are currently established for diagnosis and risk stratification. Several proteomic studies have aimed to identify PE and FGR biomarkers - often with varying results across studies. However, proteomics has revealed altered expression of human leukocyte antigen-I in PE cases, which is supported in Genome-wide association study (GWAS) studies. Proteomic results support the heterogeneous nature of PE by identification of molecular subgroups - including subgroups characterized by immune-related proteins e.g. CXCL10. No specific immunological markers are found on FGR, but differences in overall plasma proteomic signature have been suggested. SUMMARY Proteomics certainly holds great potential. The immunological component in PE and FGR are still unclarified, but improvements in proteomic technologies may provide both definition of disease subgroups and subsequent discovery of biomarkers and targeted analysis within each subgroup.
Collapse
Affiliation(s)
- Marie Winther
- Department of Clinical Immunology, the Danish National University Hospital
| | - Morten Hanefeld Dziegiel
- Department of Clinical Immunology, the Danish National University Hospital
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
47
|
Friedman AM. Placentation, Hypertensive Disorders, and Neonatal Outcomes. Paediatr Perinat Epidemiol 2025; 39:133-134. [PMID: 39961746 DOI: 10.1111/ppe.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025]
Affiliation(s)
- Alexander M Friedman
- Department of Obstetrics and Gynecology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| |
Collapse
|
48
|
Shu Z, Wang W. Predictive value of prenatal screening markers combined with serum placental growth factor in early pregnancy for preeclampsia. Pak J Med Sci 2025; 41:598-602. [PMID: 39926691 PMCID: PMC11803793 DOI: 10.12669/pjms.41.2.9794] [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: 03/08/2024] [Revised: 10/14/2024] [Accepted: 11/25/2024] [Indexed: 02/11/2025] Open
Abstract
Objective To observe the predictive value of prenatal screening markers combined with serum placental growth factor(PLGF) in early pregnancy for preeclampsia(PE). Methods This was a prospective study. A total of 369 pregnant women undergoing early pregnancy examinations were selected at Jingmen Central Hospital from August 2024 to January 2025 and divided into the PE group(n=43) and the normal group(n=326) according to the presence of PE during the follow-up. The levels of prenatal screening markers alpha-fetoprotein(AFP), serum PLGF, β-human chorionic gonadotropin(β-hCG) and pregnancy-associated plasma protein-A(PAPP-A) were compared between the two groups. Results There were 43 patients experiencing PE, with an incidence of 11.65%. The levels of PLGF, β-hCG and PAPP-A were significantly decreased in the PE group compared with those in the normal group, and the differences between the groups were statistically significant (all P<0.05). Logistic regression analysis showed that increased prenatal screening markers AFP, serum PLGF, HCG and PAPP-A were independent risk factors for PE, with statistically significant differences between the groups (all P<0.05). Finally, the results of ROC curve analysis showed that the AUCs of AFP, PLGF, β-hCG and PAPP-A were 0.618, 0.645, 0.690, and 0.645, respectively, and the AUC of combined prediction was 0.825, which was significantly increased compared with that of each marker alone, with statistically significant differences(P<0.05). Conclusion The development of PE in pregnancy is closely related to the levels of AFP, PLGF, β-hCG and PAPP-A. The predictive efficiency of combined detection of AFP, PLGF, β-hCG and PAPP-A for PE in pregnancy significantly increases.Project Title: sFlt-1/PlGF based combined early pregnancy prenatal screening marker Nursing Strategies and Application in Preeclampsia Risk Assessment, Project Number: 2024YFYB111, Year: 2024.
Collapse
Affiliation(s)
- Zitan Shu
- Zitan Shu, Department of Laboratory Medicine, Jingmen Central Hospital, Jingmen 448001, Hubei, China
| | - Weiwei Wang
- Weiwei Wang, Department of Operating Room, Jingmen Central Hospital, Jingmen 448001, Hubei, China
| |
Collapse
|
49
|
Cheng X, Gao H, Li Q, Zhang N, Lu Y. Sources of polycyclic aromatic hydrocarbons exposure and their effects on glycolipid metabolism in pregnant women and their newborn in Haikou City, China. Front Public Health 2025; 12:1510517. [PMID: 39901915 PMCID: PMC11788389 DOI: 10.3389/fpubh.2024.1510517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/31/2024] [Indexed: 02/05/2025] Open
Abstract
Background Polycyclic aromatic hydrocarbons (PAHs) are a class of large organic compounds that persist in the environment and are classified as an important subset of persistent organic pollutants (POPs). This study aimed to assess PAH exposure in pregnant women and newborns in Haikou City, China, and evaluate their impact on glycolipid metabolism. Methods A total of 300 pregnant women and their newborns were selected for the study between May 2022 and December 2023. Data on lifestyle and dietary habits were collected, and PAH levels in venous blood and umbilical cord blood were measured using gas chromatography-mass spectrometry. Glycolipid metabolism indicators, including fasting plasma glucose (FPG), triglycerides (TG), total cholesterol (TC), high-density lipoprotein (HDL), and low-density lipoprotein (LDL), were also measured. Correlation and regression analyses were conducted to explore the relationships between PAH exposure and metabolic indicators. Results Thirteen PAHs were detected in both maternal and cord blood. The median concentrations of ΣPAHs (the total concentration of the 16 detected PAHs) were 11.211 μg/L in maternal blood and 10.921 μg/L in newborns. Significant correlations were observed between PAH exposure and glycolipid metabolism, with PAH exposure linked to reduced levels of TC and TG. Factors such as proximity to roads, cooking fuel type, and dietary habits influenced PAH levels. Higher education levels were associated with lower PAH concentrations, whereas living near roads and using gas as cooking fuel increased exposure. Conclusion There is a notable risk of PAH exposure in pregnant women and newborns in Haikou, influenced by environmental and lifestyle factors. PAH exposure during pregnancy may affect glycolipid metabolism in both mothers and newborns, highlighting the need for interventions to reduce exposure.
Collapse
Affiliation(s)
- Xiaomei Cheng
- School of Public Health, Hainan Academy of Medical Sciences, Hainan Medical University, Hainan, China
| | - Haifeng Gao
- School of Public Health, Hainan Academy of Medical Sciences, Hainan Medical University, Hainan, China
| | - Qiaojun Li
- School of Public Health, Hainan Academy of Medical Sciences, Hainan Medical University, Hainan, China
| | - Naifan Zhang
- School of Public Health, Xinjiang Medical University, Xinjiang, China
| | - Ying Lu
- School of Public Health, Hainan Academy of Medical Sciences, Hainan Medical University, Hainan, China
| |
Collapse
|
50
|
Karabay G, Bayraktar B, Seyhanli Z, Filiz AA, Tokgoz Cakir B, Aktemur G, Tonyali NV, Agaoglu RT, Kocaoglu G, Karabay U, Yucel KY. Evaluation of Conventional and Combined Doppler Parameters in Preeclampsia: Diagnostic and Prognostic Insights. J Clin Med 2025; 14:647. [PMID: 39860652 PMCID: PMC11766110 DOI: 10.3390/jcm14020647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Background: The aim of this study was to examine the relationship between conventional and novel Doppler parameters, including cerebroplacental ratio (CPR), cerebral-placental-uterine ratio (CPUR), umbilical-to-cerebral ratio (UCR), and amniotic-to-umbilical-cerebral ratio (AUCR), with the diagnosis of preeclampsia (PE) and adverse neonatal outcomes in PE cases. Methods: This prospective case-control study was conducted at the Ankara Etlik City Hospital Perinatology Clinic between November 2023 and May 2024. The study population was divided into two groups: Group 1, consisting of 74 patients diagnosed with preeclampsia, and Group 2, consisting of 80 healthy control patients. Composite adverse perinatal outcomes (CANOs) include presence of at least one adverse outcome: 5th-minute APGAR score < 7, transient tachypnea of the newborn (TTN), respiratory distress syndrome (RDS), need for continuous positive airway pressure (CPAP), need for mechanical ventilation, neonatal intensive care unit (NICU) admission, neonatal hypoglycemia, need for phototherapy, intraventricular hemorrhage (IVH), and neonatal sepsis. Results: The CPR, CPUR, and AUCR were significantly lower in the PE group compared to the control group, while the UCR was notably higher in the PE group. Among the combined ratios, the CPUR exhibited the highest diagnostic performance for both PE diagnosis and the prediction of CANOs. Additionally, while the UCR, CPR, and AUCR were significant for PE diagnosis, only AUCR demonstrated a significant association with the prediction of CANOs. Conclusions: Combined Doppler parameters, especially CPUR and AUCR, offer valuable insights into diagnosing PE and predicting CANOs. CPUR demonstrated the highest diagnostic accuracy, underscoring its potential utility in clinical settings.
Collapse
Affiliation(s)
- Gulsan Karabay
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara 06170, Turkey; (B.B.); (Z.S.); (A.A.F.); (B.T.C.); (G.A.); (N.V.T.); (R.T.A.); (K.Y.Y.)
| | - Burak Bayraktar
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara 06170, Turkey; (B.B.); (Z.S.); (A.A.F.); (B.T.C.); (G.A.); (N.V.T.); (R.T.A.); (K.Y.Y.)
| | - Zeynep Seyhanli
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara 06170, Turkey; (B.B.); (Z.S.); (A.A.F.); (B.T.C.); (G.A.); (N.V.T.); (R.T.A.); (K.Y.Y.)
| | - Ahmet Arif Filiz
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara 06170, Turkey; (B.B.); (Z.S.); (A.A.F.); (B.T.C.); (G.A.); (N.V.T.); (R.T.A.); (K.Y.Y.)
| | - Betul Tokgoz Cakir
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara 06170, Turkey; (B.B.); (Z.S.); (A.A.F.); (B.T.C.); (G.A.); (N.V.T.); (R.T.A.); (K.Y.Y.)
| | - Gizem Aktemur
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara 06170, Turkey; (B.B.); (Z.S.); (A.A.F.); (B.T.C.); (G.A.); (N.V.T.); (R.T.A.); (K.Y.Y.)
| | - Nazan Vanli Tonyali
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara 06170, Turkey; (B.B.); (Z.S.); (A.A.F.); (B.T.C.); (G.A.); (N.V.T.); (R.T.A.); (K.Y.Y.)
| | - Recep Taha Agaoglu
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara 06170, Turkey; (B.B.); (Z.S.); (A.A.F.); (B.T.C.); (G.A.); (N.V.T.); (R.T.A.); (K.Y.Y.)
| | - Gulcan Kocaoglu
- Department of Obstetrics and Gynecology, Ankara Etlik City Hospital, Ankara 06170, Turkey;
| | - Umut Karabay
- Department of Internal Medicine, Gulhane Training and Research Hospital, Ankara 06010, Turkey;
| | - Kadriye Yakut Yucel
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara 06170, Turkey; (B.B.); (Z.S.); (A.A.F.); (B.T.C.); (G.A.); (N.V.T.); (R.T.A.); (K.Y.Y.)
| |
Collapse
|