1
|
Li Y, Yan X, Yu H, Zhou Y, Gao Y, Zhou X, Yuan Y, Ding Y, Shi Q, Fang Y, Du H, Yuan E, Zhao X, Zhang L. Downregulation of CMIP contributes to preeclampsia development by impairing trophoblast function via the PDE7B-cAMP pathway. Cell Mol Life Sci 2025; 82:203. [PMID: 40372501 PMCID: PMC12081820 DOI: 10.1007/s00018-025-05726-5] [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: 01/20/2025] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Preeclampsia (PE) is one of the leading causes of perinatal maternal and fetal morbidity and mortality, but its precise mechanism remains elusive. Previous research has suggested that c-Maf-inducible protein (CMIP) is abnormally expressed in PE pathophysiology. Therefore, we aimed to explore the potential role of CMIP and its downstream molecules in PE. METHODS Multiplex immunofluorescence and immunohistochemical assays were conducted on preeclamptic placentas. Functional analysis of CMIP was performed in HTR-8/SVneo cells through transfection experiments in which either CMIP was overexpressed or downregulated. RNA sequencing was utilized to identify the molecular pathways downstream of CMIP. The impact of hypoxia on CMIP levels was assessed in three different types of trophoblast cells. The therapeutic efficacy of CMIP was evaluated in an N(ω)-nitro-L-arginine methyl ester (L-NAME)-induced rat model of PE. RESULTS CMIP expression was downregulated in extrachorionic trophoblasts (EVTs) and syncytiotrophoblasts (STBs) in preeclamptic placentas. This downregulation of CMIP in trophoblast cells disrupts cell proliferation, migration, invasion, and angiogenesis by upregulating the PDE7B-cAMP pathway, while elevated CMIP levels enhance these cellular functions. Hypoxia reduced CMIP expression in all three types of trophoblast cells. Moreover, in a rat model of PE, supplementation with CMIP alleviated hypertension and increased fetal weight and number. CONCLUSIONS Our study demonstrates for the first time that the CMIP-PDE7B-cAMP pathway contributes to PE development by influencing trophoblast function. The signaling pathway proteins involved in PE induced by CMIP may provide new clues to the occurrence of PE and new targets for future PE therapy.
Collapse
Affiliation(s)
- Yina Li
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, 7 Kangfu Qian Street, Zhengzhou, Henan, 450052, People's Republic of China
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, China
| | - Xinjing Yan
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, 7 Kangfu Qian Street, Zhengzhou, Henan, 450052, People's Republic of China
- Department of Blood Transfusion, Xi'an People's Hospital, Xi'an, 710000, China
| | - Haiyang Yu
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, 7 Kangfu Qian Street, Zhengzhou, Henan, 450052, People's Republic of China
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, China
| | - Yuanbo Zhou
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, 7 Kangfu Qian Street, Zhengzhou, Henan, 450052, People's Republic of China
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, China
| | - Yongrui Gao
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, 7 Kangfu Qian Street, Zhengzhou, Henan, 450052, People's Republic of China
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, China
| | - Xinyuan Zhou
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, 7 Kangfu Qian Street, Zhengzhou, Henan, 450052, People's Republic of China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujie Yuan
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, 7 Kangfu Qian Street, Zhengzhou, Henan, 450052, People's Republic of China
| | - Yangnan Ding
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, 7 Kangfu Qian Street, Zhengzhou, Henan, 450052, People's Republic of China
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, China
| | - Qianqian Shi
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, 7 Kangfu Qian Street, Zhengzhou, Henan, 450052, People's Republic of China
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, China
| | - Yang Fang
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, 7 Kangfu Qian Street, Zhengzhou, Henan, 450052, People's Republic of China
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, China
| | - Hongmei Du
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, 7 Kangfu Qian Street, Zhengzhou, Henan, 450052, People's Republic of China
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, China
| | - Enwu Yuan
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, 7 Kangfu Qian Street, Zhengzhou, Henan, 450052, People's Republic of China
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, China
- Tianjian Advanced Biomedical Laboratory, Zhengzhou, China
| | - Xin Zhao
- Tianjian Advanced Biomedical Laboratory, Zhengzhou, China.
- Radiology Department, Third Affiliated Hospital of Zhengzhou University, 7 Kangfu Qian Street, Zhengzhou, Henan, 450052, People's Republic of China.
| | - Linlin Zhang
- Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, 7 Kangfu Qian Street, Zhengzhou, Henan, 450052, People's Republic of China.
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, China.
- Tianjian Advanced Biomedical Laboratory, Zhengzhou, China.
| |
Collapse
|
2
|
Zhao Z, Liu X, Guan Y, Li C, Wang Z. Exploring the potential of cell-free RNA and Pyramid Scene Parsing Network for early preeclampsia screening. BMC Pregnancy Childbirth 2025; 25:445. [PMID: 40229739 PMCID: PMC11995606 DOI: 10.1186/s12884-025-07503-5] [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: 12/10/2024] [Accepted: 03/20/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Circulating cell-free RNA (cfRNA) is gaining recognition as an effective biomarker for the early detection of preeclampsia (PE). However, the current methods for selecting disease-specific biomarkers are often inefficient and typically one-dimensional. PURPOSE This study introduces a Pyramid Scene Parsing Network (PSPNet) model to predict PE, aiming to improve early risk assessment using cfRNA profiles. METHODS The theoretical maximum Preeclamptic Risk Index (PRI) of patients clinically diagnosed with PE is defined as "1", and the control group (NP) is defined as "0", referred to as the clinical PRI. A data preprocessing algorithm was used to screen relevant cfRNA indicators for PE. The cfRNA expression profiles were obtained from the Gene Expression Omnibus (GSE192902), consisting of 180 normal pregnancies (NP) and 69 preeclamptic (PE) samples, collected at two gestational time points: ≤ 12 weeks and 13-20 weeks. Based on the differences in cfRNA expression profiles, the Calculated Ground Truth values of the NP and PE groups in the sequencing data were acquired (Calculated PRI). The differential algorithm was embedded in the PSPNet neural network and the network was then trained using the generated dataset. Subsequently, the real-world sequencing dataset was used to validate and optimize the network, ultimately outputting the PRI values of the healthy control group and the PE group (PSPNet-based PRI). The model's predictive ability for PE was evaluated by comparing the fit between Calculated PRI (Calculated Ground Truth) and PSPNet-based PRI. RESULTS The mean absolute error (MAE) between the Calculated Ground Truth the PSPNet-based PRI was 0.0178 for cfRNA data sampled at ≤ 12 gws and 0.0195 for data sampled at 13-20 gws. For cfRNA data sequenced at ≤ 12 gws and 13-20 gws, the corresponding loss values, maximum absolute errors, peak-to-valley error values, mean absolute errors, and average prediction times per sample were 0.0178 (0.0195). CONCLUSIONS The present PSPNet model is reliable and fast for cfRNA-based PE prediction and its PRI output allows for continuous PE risk monitoring, introducing an innovative and effective method for early PE prediction. This model enables timely interventions and better management of pregnancy complications, particularly benefiting densely populated developing countries with high PE incidence and limited access to routine prenatal care.
Collapse
Affiliation(s)
- Zhuo Zhao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, No.98, Xiwu Road, Xi'an, Shaanxi, People's Republic of China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, No.98, Xiwu Road, Xi'an, Shaanxi, People's Republic of China
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoxu Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xian Jiaotong University, Xi'an, China
| | - Yonghui Guan
- Department of Urology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Chunfang Li
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, No.76, Yanta West Road, Xi'an, Shaanxi, People's Republic of China.
| | - Zheng Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, No.98, Xiwu Road, Xi'an, Shaanxi, People's Republic of China.
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, No.98, Xiwu Road, Xi'an, Shaanxi, People's Republic of China.
| |
Collapse
|
3
|
Zhang T, Li H, Jiang E, Zhang L, Liu L, Zhang C. CSPG4 involvement in endometrial decidualization contributes to the pathogenesis of preeclampsia†. Biol Reprod 2025; 112:361-374. [PMID: 39563514 DOI: 10.1093/biolre/ioae167] [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: 07/30/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 11/21/2024] Open
Abstract
Preeclampsia (PE) is a condition of pregnancy in which symptoms of hypertension develop after 20 weeks of gestation. it can lead to placental dysfunction, maternal and perinatal mortality and morbidity. The incidence of PE is increasing, posing a serious threat to the lives of pregnant women and their unborn children. Currently, most of the research on the pathogenesis of PE has focused on placenta, However, maternal decidualization is the basis for placental formation and growth. Chondroitin sulfate proteoglycan 4 (CSPG4) is a transmembrane protein that plays a role in cell proliferation, invasion, and migration. However, its function during decidualization is not yet understood. In this study, we investigated the role of CSPG4 and found that its expression was significantly down-regulated in the decidual tissue of patients with severe PE compared to normal pregnant women. During artificially induced decidualization, CSPG4 expression was significantly increased. Knockdown of CSPG4 by small interfering RNA inhibited decidualization, which, in turn, inhibited the invasion of trophoblast cells. In both pseudopregnant and pregnant mice, endometrial stromal cells proliferated rapidly and Cspg4 expression increased during decidualization. Therefore, we believe that CSPG4 plays a crucial role in the process of decidualization. The defect in decidualization caused by abnormal CSPG4 expression could lead to insufficient trophoblast invasion, ultimately contributing to the occurrence of PE.
Collapse
Affiliation(s)
- Tianying Zhang
- Department of Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
- Taishan Polytechnic, Taian, Shandong 271000, China
| | - Hua Li
- Department of Gynecology and Obstetrics, Ji'nan Maternity and Child Care Hospital, Jinan, Shandong, China
| | - Enhui Jiang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Liang Zhang
- Research Center of Translational Medicine, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Lisheng Liu
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Cong Zhang
- Department of Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| |
Collapse
|
4
|
Nguyen-Hoang L, Dinh LT, Tai AS, Nguyen DA, Pooh RK, Shiozaki A, Zheng M, Hu Y, Li B, Kusuma A, Yapan P, Gosavi A, Kaneko M, Luewan S, Chang TY, Chaiyasit N, Nanthakomon T, Liu H, Shaw SW, Leung WC, Mahdy ZA, Aguilar A, Leung HH, Lee NM, Lau SL, Wah IY, Lu X, Sahota DS, Chong MK, Poon LC. Implementation of First-Trimester Screening and Prevention of Preeclampsia: A Stepped Wedge Cluster-Randomized Trial in Asia. Circulation 2024; 150:1223-1235. [PMID: 38923439 PMCID: PMC11472904 DOI: 10.1161/circulationaha.124.069907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND This trial aimed to assess the efficacy, acceptability, and safety of a first-trimester screen-and-prevent strategy for preterm preeclampsia in Asia. METHODS Between August 1, 2019, and February 28, 2022, this multicenter stepped wedge cluster randomized trial included maternity/diagnostic units from 10 regions in Asia. The trial started with a period where all recruiting centers provided routine antenatal care without study-related intervention. At regular 6-week intervals, one cluster was randomized to transit from nonintervention phase to intervention phase. In the intervention phase, women underwent first-trimester screening for preterm preeclampsia using a Bayes theorem-based triple-test. High-risk women, with adjusted risk for preterm preeclampsia ≥1 in 100, received low-dose aspirin from <16 weeks until 36 weeks. RESULTS Overall, 88.04% (42 897 of 48 725) of women agreed to undergo first-trimester screening for preterm preeclampsia. Among those identified as high-risk in the intervention phase, 82.39% (2919 of 3543) received aspirin prophylaxis. There was no significant difference in the incidence of preterm preeclampsia between the intervention and non-intervention phases (adjusted odds ratio [aOR], 1.59 [95% CI, 0.91-2.77]). However, among high-risk women in the intervention phase, aspirin prophylaxis was significantly associated with a 41% reduction in the incidence of preterm preeclampsia (aOR, 0.59 [95% CI, 0.37-0.92]). In addition, it correlated with 54%, 55%, and 64% reduction in the incidence of preeclampsia with delivery at <34 weeks (aOR, 0.46 [95% CI, 0.23-0.93]), spontaneous preterm birth <34 weeks (aOR, 0.45 [95% CI, 0.22-0.92]), and perinatal death (aOR, 0.34 [95% CI, 0.12-0.91]), respectively. There was no significant between-group difference in the incidence of aspirin-related severe adverse events. CONCLUSIONS The implementation of the screen-and-prevent strategy for preterm preeclampsia is not associated with a significant reduction in the incidence of preterm preeclampsia. However, low-dose aspirin effectively reduces the incidence of preterm preeclampsia by 41% among high-risk women. The screen-and-prevent strategy for preterm preeclampsia is highly accepted by a diverse group of women from various ethnic backgrounds beyond the original population where the strategy was developed. These findings underpin the importance of the widespread implementation of the screen-and-prevent strategy for preterm preeclampsia on a global scale. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT03941886.
Collapse
Affiliation(s)
- Long Nguyen-Hoang
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital (L.N.-H., A.S.T.T., H.H.Y.L., N.M.W.L., S.L.L., I.Y.M.W., X.L., D.S.S., L.C.P.), Chinese University of Hong Kong
| | - Linh Thuy Dinh
- Center for Prenatal and Neonatal Screening and Diagnosis, Hanoi Obstetrics and Gynecology Hospital, Vietnam (L.T.D., D.-A.N.)
| | - Angela S.T. Tai
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital (L.N.-H., A.S.T.T., H.H.Y.L., N.M.W.L., S.L.L., I.Y.M.W., X.L., D.S.S., L.C.P.), Chinese University of Hong Kong
| | - Duy-Anh Nguyen
- Center for Prenatal and Neonatal Screening and Diagnosis, Hanoi Obstetrics and Gynecology Hospital, Vietnam (L.T.D., D.-A.N.)
| | - Ritsuko K. Pooh
- Clinical Research Institute of Fetal Medicine Prenatal Medical Clinic, Osaka, Japan (R.K.P.)
| | - Arihiro Shiozaki
- Department of Obstetrics and Gynecology, Toyama University Hospital, Toyama, Japan (A.S.)
| | - Mingming Zheng
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital Affiliated to Nanjing University Medical School, China (M.Z., Y.H.)
| | - Yali Hu
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital Affiliated to Nanjing University Medical School, China (M.Z., Y.H.)
| | - Bin Li
- Department of Obstetrics and Gynecology, Kunming Angel Women and Children’s Hospital, Teaching Hospital of Kunming University of Science and Technology, China (B.L.)
| | - Aditya Kusuma
- Department of Obstetrics and Gynecology, Harapan Kita Women and Children Hospital, Jakarta, Indonesia (A.K.)
| | - Piengbulan Yapan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Siriraj Hospital, Bangkok, Thailand (P.Y.)
| | - Arundhati Gosavi
- Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore (A.G.)
| | - Mayumi Kaneko
- Department of Obstetrics and Gynecology, Showa University Hospital, Tokyo, Japan (M.K.)
| | - Suchaya Luewan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Chiang Mai University, Thailand (S.L.)
| | - Tung-Yao Chang
- Department of Fetal Medicine, Taiji Clinic, Taipei, Taiwan (T.-Y.C.)
| | - Noppadol Chaiyasit
- Department of Obstetrics and Gynecology, King Chulalongkorn Memorial Hospital, Bangkok, Thailand (N.C.)
| | - Tongta Nanthakomon
- Department of Obstetrics and Gynecology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand (T.N.)
| | - Huishu Liu
- Department of Obstetrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, China (H.L.)
| | - Steven W. Shaw
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taiwan (S.W.S.)
| | - Wing Cheong Leung
- Department of Obstetrics and Gynaecology, Kwong Wah Hospital, Hong Kong SAR, China (W.C.L.)
| | - Zaleha Abdullah Mahdy
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia (Z.A.M.)
| | - Angela Aguilar
- Department of Obstetrics and Gynecology, University of the Philippines College of Medicine, Philippine General Hospital, Manila (A.A.)
| | - Hillary H.Y. Leung
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital (L.N.-H., A.S.T.T., H.H.Y.L., N.M.W.L., S.L.L., I.Y.M.W., X.L., D.S.S., L.C.P.), Chinese University of Hong Kong
| | - Nikki M.W. Lee
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital (L.N.-H., A.S.T.T., H.H.Y.L., N.M.W.L., S.L.L., I.Y.M.W., X.L., D.S.S., L.C.P.), Chinese University of Hong Kong
| | - So Ling Lau
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital (L.N.-H., A.S.T.T., H.H.Y.L., N.M.W.L., S.L.L., I.Y.M.W., X.L., D.S.S., L.C.P.), Chinese University of Hong Kong
| | - Isabella Y.M. Wah
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital (L.N.-H., A.S.T.T., H.H.Y.L., N.M.W.L., S.L.L., I.Y.M.W., X.L., D.S.S., L.C.P.), Chinese University of Hong Kong
| | - Xiaohong Lu
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital (L.N.-H., A.S.T.T., H.H.Y.L., N.M.W.L., S.L.L., I.Y.M.W., X.L., D.S.S., L.C.P.), Chinese University of Hong Kong
| | - Daljit S. Sahota
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital (L.N.-H., A.S.T.T., H.H.Y.L., N.M.W.L., S.L.L., I.Y.M.W., X.L., D.S.S., L.C.P.), Chinese University of Hong Kong
| | - Marc K.C. Chong
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine (M.K.C.C.), Chinese University of Hong Kong
| | - Liona C. Poon
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital (L.N.-H., A.S.T.T., H.H.Y.L., N.M.W.L., S.L.L., I.Y.M.W., X.L., D.S.S., L.C.P.), Chinese University of Hong Kong
| |
Collapse
|
5
|
Deepak V, El-Balawi L, Harris LK. Placental Drug Delivery to Treat Pre-Eclampsia and Fetal Growth Restriction. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2311165. [PMID: 38745536 DOI: 10.1002/smll.202311165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 04/23/2024] [Indexed: 05/16/2024]
Abstract
Pre-eclampsia and fetal growth restriction (FGR) continue to cause unacceptably high levels of morbidity and mortality, despite significant pharmaceutical and technological advances in other disease areas. The recent pandemic has also impacted obstetric care, as COVID-19 infection increases the risk of poor pregnancy outcomes. This review explores the reasons why it lacks effective drug treatments for the placental dysfunction that underlies many common obstetric conditions and describes how nanomedicines and targeted drug delivery approaches may provide the solution to the current drug drought. The ever-increasing range of biocompatible nanoparticle formulations available is now making it possible to selectively deliver drugs to uterine and placental tissues and dramatically limit fetal drug transfer. Formulations that are refractory to placental uptake offer the possibility of retaining drugs within the maternal circulation, allowing pregnant individuals to take medicines previously considered too harmful to the developing baby. Liposomes, ionizable lipid nanoparticles, polymeric nanoparticles, and adenoviral vectors have all been used to create efficacious drug delivery systems for use in pregnancy, although each approach offers distinct advantages and limitations. It is imperative that recent advances continue to be built upon and that there is an overdue investment of intellectual and financial capital in this field.
Collapse
Affiliation(s)
- Venkataraman Deepak
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9WL, UK
- St Mary's Hospital, Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| | - Lujain El-Balawi
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL, UK
| | - Lynda K Harris
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9WL, UK
- St Mary's Hospital, Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL, UK
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
6
|
Sun L, He Y, Chen J, Yang X, Ding Y, Shi M, He A, Zhang P, Huang Z, Li R. Bioinformatics analysis identifies potential autophagy key genes and immune infiltration in preeclampsia. J Obstet Gynaecol Res 2024; 50:618-632. [PMID: 38350492 DOI: 10.1111/jog.15902] [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: 09/27/2023] [Accepted: 01/30/2024] [Indexed: 02/15/2024]
Abstract
BACKGROUND Preeclampsia (PE) is a disease that seriously threatens maternal and fetal health. Appropriate autophagy can shield the placenta from oxidative stress, but its role in PE is unclear. OBJECTIVE To identify potential autophagy-related genes in PE. METHODS Microarray datasets from the Gene Expression Omnibus database, compassing the test dataset GSE10588, along with validation datasets GSE4707 and GSE60438 GPL10558, were utilized. Differentially expressed genes (DEGs) were identified using the limma R package, intersected with autophagy-related genes. Hub genes were obtained using the Cytoscape software and analyzed via gene set enrichment analysis (GSEA). The diagnostic capability of hub genes was evaluated using receiver operating characteristic (ROC) curve analysis. Analysis of immune cell infiltration was conducted using single-sample gene set enrichment analysis (ssGSEA) and CIBERSORT methods. Placental tissues were collected from 10 normal pregnant women and 10 preeclamptic pregnant women, and the expression of hub genes was validated through immunohistochemistry and western blot analysis. RESULTS Analysis of the microarray data identified 2224 DEGs, among which 26 were autophagy-related DEGs identified through intersection with autophagy genes. Ten hub genes were identified. Immune cell infiltration analysis suggested the potential involvement of T regulatory cells (Tregs), natural killer cells, neutrophils, and T follicular helper cells in the pathogenesis of PE. ROC curve analysis indicated promising diagnostic capabilities for EGFR and TP53. Additionally, levels of EGFR and TP53 were significantly higher in placental tissue from PE pregnancies compared to normal pregnancies. CONCLUSION EGFR and TP53 may play a role in PE by influencing autophagy.
Collapse
Affiliation(s)
- Lu Sun
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yanhong He
- Department of Obstetrics and Gynecology, The Affiliated Shunde hospital of Jinan University, the Second People's Hospital of Shunde, Foshan, China
| | - Jie Chen
- Department of Obstetrics and Gynecology, The Affiliated Shunde hospital of Jinan University, the Second People's Hospital of Shunde, Foshan, China
| | - Xiaofeng Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yuzhen Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Meiting Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Andong He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ping Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhengrui Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ruiman Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
7
|
Chen J, Huai J, Yang H. Low-molecular-weight heparin for the prevention of preeclampsia in high-risk pregnancies without thrombophilia: a systematic review and meta-analysis. BMC Pregnancy Childbirth 2024; 24:68. [PMID: 38233773 PMCID: PMC10792962 DOI: 10.1186/s12884-023-06218-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 12/20/2023] [Indexed: 01/19/2024] Open
Abstract
OBJECTIVES To systematically evaluate the efficacy of low molecular weight heparin (LMWH) to prevent preeclampsia in high risk pregnant women without thrombophilia. SEARCH STRATEGY PubMed, Embase and the Cochrane library were searched for articles published before 1st August 2022 using the combination keywords "preeclampsia", "Low Molecular Weight Heparin", "LMWH", "Heparin, Low Molecular Weight", "Dalteparin", "Nadroparin", and "Tinzaparin". SELECTION CRITERIA Randomized controlled trials evaluating the use of LMWH in pregnant women at high risk of preeclampsia without thrombophilia. DATA COLLECTION AND ANALYSIS Ten studies were included in the meta-analysis (1758 patients in total). Outcomes were expressed as relative risk (RR) with 95% confidence intervals (CI). RESULTS LMWH reduced the incidence of PE (RR = 0.67; 95% CI = 0.50-0.90; P = 0.009) in high risk pregnant women without thrombophilia. Subgroup analysis found that the prophylactic effect of LMWH was only significant in studies using low-dose aspirin (LDA) as the primary intervention. The combination of LMWH and LDA was also effective for the prevention of preterm birth and fetal growth restriction, but had no effect on the incidence of placenta abruption. CONCLUSION For women at high risk of developing preeclampsia without thrombophilia, the combination of LMWH and low-dose aspirin is effective for the prevention of preeclampsia, preterm birth and fetal growth restriction and is superior to LDA alone.
Collapse
Affiliation(s)
- Jiahui Chen
- Department of Obstetrics and Gynecology, Peking University First Hospital, No. 1 Xi 'an Men Street, Xicheng District, Beijing, 100034, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Jing Huai
- Department of Obstetrics and Gynecology, Peking University First Hospital, No. 1 Xi 'an Men Street, Xicheng District, Beijing, 100034, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Huixia Yang
- Department of Obstetrics and Gynecology, Peking University First Hospital, No. 1 Xi 'an Men Street, Xicheng District, Beijing, 100034, China.
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China.
| |
Collapse
|
8
|
Reeder HT, Haneuse S, Modest AM, Hacker MR, Sudhof LS, Papatheodorou SI. A novel approach to joint prediction of preeclampsia and delivery timing using semicompeting risks. Am J Obstet Gynecol 2023; 228:338.e1-338.e12. [PMID: 36037998 PMCID: PMC9968360 DOI: 10.1016/j.ajog.2022.08.045] [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: 07/24/2022] [Revised: 08/20/2022] [Accepted: 08/20/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Preeclampsia is a pregnancy complication that contributes substantially to perinatal morbidity and mortality worldwide. Existing approaches to modeling and prediction of preeclampsia typically focus either on predicting preeclampsia risk alone, or on the timing of delivery following a diagnosis of preeclampsia. As such, they are misaligned with typical healthcare interactions during which the 2 events are generally considered simultaneously. OBJECTIVE This study aimed to describe the "semicompeting risks" framework as an innovative approach for jointly modeling the risk and timing of preeclampsia and the timing of delivery simultaneously. Through this approach, one can obtain, at any point during the pregnancy, clinically relevant summaries of an individual's predicted outcome trajectories in 4 risk categories: not developing preeclampsia and not having delivered, not developing preeclampsia but having delivered because of other causes, developing preeclampsia but not having delivered, and developing preeclampsia and having delivered. STUDY DESIGN To illustrate the semicompeting risks methodology, we presented an example analysis of a pregnancy cohort from the electronic health record of an urban, academic medical center in Boston, Massachusetts (n=9161 pregnancies). We fit an illness-death model with proportional-hazards regression specifications describing 3 hazards for timings of preeclampsia, delivery in the absence of preeclampsia, and delivery following preeclampsia diagnosis. RESULTS The results indicated nuanced relationships between a variety of risk factors and the timings of preeclampsia diagnosis and delivery, including maternal age, race/ethnicity, parity, body mass index, diabetes mellitus, chronic hypertension, cigarette use, and proteinuria at 20 weeks' gestation. Sample predictions for a diverse set of individuals highlighted differences in projected outcome trajectories with regard to preeclampsia risk and timing, and timing of delivery either before or after preeclampsia diagnosis. CONCLUSION The semicompeting risks framework enables characterization of the joint risk and timing of preeclampsia and delivery, providing enhanced, meaningful information regarding clinical decision-making throughout the pregnancy.
Collapse
Affiliation(s)
- Harrison T Reeder
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Sebastien Haneuse
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Anna M Modest
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA; Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA
| | - Michele R Hacker
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA; Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Leanna S Sudhof
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA; Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA
| | | |
Collapse
|
9
|
Akbari R, Hantoushzadeh S, Panahi Z, Bahonar S, Ghaemi M. A bibliometric review of 35 years of studies about preeclampsia. Front Physiol 2023; 14:1110399. [PMID: 36818438 PMCID: PMC9932928 DOI: 10.3389/fphys.2023.1110399] [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: 11/28/2022] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
The purpose of this study is to investigate preeclampsia. It used the visualization tools of CiteSpace, VOSviewer, Gunnmap, Bibliometrix®, and Carrot2 to analyze 3,754 preeclampsia studies from 1985 to 2020 in Obstetrics and Gynecology areas. Carrot2 was used to explain each cluster in extra detail. The results found that there is an increasing trend in many publications related to preeclampsia from 1985 to 2020. The number of studies on preeclampsia has increased significantly in the last century. Analysis of the keywords found a strong relationship with preeclampsia concepts and keywords classified into five categories. Co-citation analysis was also performed which was classified into six categories. Reading the article offers important to support not only to grind the context of preeclampsia challenges but also to design a new trend in this field. The number of studies on preeclampsia has substantially improved over the decades ago. The findings of documents published from 1985 to 2020 showed three stages in research on this subject: 1985 to 1997 (a seeding stage), 1997-2005 (rapid growth stage), and 2005 onwards (development stage).
Collapse
Affiliation(s)
- Razieh Akbari
- School of Medicine, Department of Obstetrics and Gynecology, Tehran University of Medical Sciences, Tehran, Iran
| | - Sedigheh Hantoushzadeh
- School of Medicine, Department of Obstetrics and Gynecology, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Panahi
- School of Medicine, Department of Obstetrics and Gynecology, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Marjan Ghaemi
- School of Medicine, Department of Obstetrics and Gynecology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Rocha G. Consequences of early-onset preeclampsia on neonatal morbidity and mortality. Minerva Pediatr (Torino) 2023; 75:87-97. [PMID: 35373936 DOI: 10.23736/s2724-5276.22.06714-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Preterm birth is the leading cause of perinatal morbidity and mortality in developed countries. Common reasons for indicated preterm births include pre-eclampsia. The increase in incidences of morbidity and mortality observed in neonates resulting from pregnancies complicated by preeclampsia is also due to alterations in angiogenic and pro-inflammatory factors that directly affect the neonatal health. This review was prepared with the aim of gathering the information available at PubMed/MEDLINE, in the years from 2011 to 2021, on the consequences of neonatal morbidity and mortality of early-onset preeclampsia. There is great controversy in the literature and paucity of studies. Early onset pre-eclampsia has been linked to fetal growth restriction (FGR). Most studies support its association with respiratory distress syndrome (RDS). Most studies point to an association between preeclampsia and bronchopulmonary dysplasia (BPD), with the highest risk in FGR. The association between preeclampsia, patent ductus arteriosus (PDA) and sepsis is not supported by the literature. The association to necrotizing enterocolitis (NEC) is controversial. The risk of spontaneous intestinal perforation (SIP) seems to be increased with preeclampsia. The association between intraventricular hemorrhage (IVH) and preeclampsia is controversial, however, preeclampsia seems to have a protective effect on periventricular leukomalacia (PVL). Most of the evidence points to the non-association between preeclampsia and retinopathy of prematurity (ROP). Hematological changes such as neutropenia, thrombocytopenia and increased nucleated red blood cell counts have been shown to be associated with preeclampsia. The evidence is still quite controversial regarding mortality. The early installation of preeclampsia will have direct consequences on neonatal morbidity. Gestational age at preterm birth is the main risk factor on neonatal morbidity. Obstetricians should aim to prolong the pregnancies complicated by early-onset severe preeclampsia as far as maternal conditions allow. This policy may contribute to improve the neonatal outcomes.
Collapse
Affiliation(s)
- Gustavo Rocha
- Department of Neonatology, São João University Hospital, Porto, Portugal -
| |
Collapse
|
11
|
Wang H, Shi Y, Ma J, Wang W, Gao J, Zhao L, Zhao T, Ding G. Integrated Proteomic and N-Glycoproteomic Profiling of Placental Tissues of Patients with Preeclampsia. Int J Womens Health 2023; 15:59-68. [PMID: 36660462 PMCID: PMC9844819 DOI: 10.2147/ijwh.s387672] [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: 08/26/2022] [Accepted: 12/13/2022] [Indexed: 01/15/2023] Open
Abstract
Background Preeclampsia (PE) is a multi-system disorder of pregnancy that poses a serious threat to maternal and perinatal health worldwide. This study aims to evaluate the global alterations of protein expression and N-glycosylations that are crucial for PE pathogenesis. Here, tandem mass tag labeling combined with LC-MS/MS was employed to determine the global expression of all proteins and intact glycopeptide in placentas from three healthy pregnant women, three patients with early-onset severe PE, and three patients with late-onset severe PE. Results A total of 2260 proteins were quantified across 9 placental tissues, of which 37 and 23 were differentially expressed in the early-onset and late-onset PE groups, compared to the controls. A total of 789 glycopeptides were accurately quantified, which were derived from 204 glycosylated sites in 159 glycoproteins and were modified by 59 N-Linked glycans. A total of 123 differently expressed glycopeptides, which were from 47 glycoproteins were identified among three groups. Through a combined analysis of proteomic and glycoproteomic data, it was found that the changes in 10 glycoproteins were caused by the difference in glycosylation level but not in the protein abundance level. Conclusion This is the first study to conduct an integrated proteomic and glycoproteomic characterization of placental tissues of PE patients. Our findings suggest that glycosylation modification may affect the molecular function of proteins through changes in the glycosylation structure or the occupancy of glycosylation, which will provide new insights to help elucidating the pathogenic mechanism of PE.
Collapse
Affiliation(s)
- Huijuan Wang
- The National Engineering Research Center for Miniaturized Detection Systems, Northwest University, Xi’an, Shaanxi, 710069, People’s Republic of China
| | - Yinmin Shi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an, Shaanxi, 710069, People’s Republic of China
| | - Jiying Ma
- The National Engineering Research Center for Miniaturized Detection Systems, Northwest University, Xi’an, Shaanxi, 710069, People’s Republic of China
| | - Wenxia Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an, Shaanxi, 710069, People’s Republic of China
| | - Jianrong Gao
- Department of Obstetrics, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, 710018, People’s Republic of China
| | - Lili Zhao
- The National Engineering Research Center for Miniaturized Detection Systems, Northwest University, Xi’an, Shaanxi, 710069, People’s Republic of China
| | - Ting Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an, Shaanxi, 710069, People’s Republic of China
| | - Guifeng Ding
- Department of Obstetrics and Gynecology, Urumqi Maternal and Child Health Care Hospital, Urumqi, Xinjiang, 830001, People’s Republic of China,Correspondence: Guifeng Ding, Department of Obstetrics and Gynecology, Urumqi Maternal and Child Health Care Hospital, No. 344 Jiefang South Road, Urumqi, Xinjiang, People’s Republic of China, Tel +86 1 331 988 0258, Fax + 86 991-8554656, Email
| |
Collapse
|
12
|
Zhang H, Li X, Zhang T, Zhou Q, Zhang C. Establishment and validation of a predictive model of preeclampsia based on transcriptional signatures of 43 genes in decidua basalis and peripheral blood. BMC Bioinformatics 2022; 23:527. [PMID: 36476092 PMCID: PMC9730617 DOI: 10.1186/s12859-022-05086-y] [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: 04/30/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Preeclampsia (PE) has an increasing incidence worldwide, and there is no gold standard for prediction. Recent progress has shown that abnormal decidualization and impaired vascular remodeling are essential to PE pathogenesis. Therefore, it is of great significance to analyze the decidua basalis and blood changes of PE to explore new methods. Here, we performed weighted gene co-expression network analysis based on 9553 differentially expressed genes of decidua basalis data (GSE60438 includes 25 cases of PE and 23 non-cases) from Gene Expression Omnibus to screen relevant module-eigengenes (MEs). Among them, MEblue and MEgrey are the most correlated with PE, which contains 371 core genes. Subsequently, we applied the logistic least absolute shrinkage and selection operator regression, screened 43 genes most relevant to prediction from the intersections of the 371 genes and training set (GSE48424 includes 18 cases of PE and 18 non-cases) genes, and built a predictive model. The specificity and sensitivity are illustrated by receiver operating characteristic curves, and the stability was verified by two validation sets (GSE86200 includes 12 cases of PE and 48 non-cases, and GSE85307 includes 47 cases of PE and 110 non-cases). The results demonstrated that our predictive model shows good predictions, with an area under the curve of 0.991 for the training set, 0.874 and 0.986 for the validation sets. Finally, we found the 43 key marker genes in the model are closely associated with the clinically accepted predictive molecules, including FLT1, PIGF, ENG and VEGF. Therefore, this predictive model provides a potential approach for PE diagnosis and treatment.
Collapse
Affiliation(s)
- Hongya Zhang
- grid.16821.3c0000 0004 0368 8293Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135 China ,grid.410585.d0000 0001 0495 1805Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 88 East Wenhua Road, Jinan, 250014 Shandong China ,grid.452927.f0000 0000 9684 550XShanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135 China
| | - Xuexiang Li
- grid.410585.d0000 0001 0495 1805Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 88 East Wenhua Road, Jinan, 250014 Shandong China
| | - Tianying Zhang
- grid.410585.d0000 0001 0495 1805Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 88 East Wenhua Road, Jinan, 250014 Shandong China
| | - Qianhui Zhou
- grid.410585.d0000 0001 0495 1805Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 88 East Wenhua Road, Jinan, 250014 Shandong China
| | - Cong Zhang
- grid.16821.3c0000 0004 0368 8293Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135 China ,grid.410585.d0000 0001 0495 1805Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 88 East Wenhua Road, Jinan, 250014 Shandong China ,grid.452927.f0000 0000 9684 550XShanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135 China
| |
Collapse
|
13
|
Kondoh K, Akahori H, Muto Y, Terada T. Identification of Key Genes and Pathways Associated with Preeclampsia by a WGCNA and an Evolutionary Approach. Genes (Basel) 2022; 13:genes13112134. [PMID: 36421809 PMCID: PMC9690438 DOI: 10.3390/genes13112134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/04/2022] [Accepted: 11/09/2022] [Indexed: 11/18/2022] Open
Abstract
Preeclampsia (PE) is the serious obstetric-related disease characterized by newly onset hypertension and causes damage to the kidneys, brain, liver, and more. To investigate genes with key roles in PE’s pathogenesis and their contributions, we used a microarray dataset of normotensive and PE patients and conducted a weighted gene co-expression network analysis (WGCNA). Cyan and magenta modules that are highly enriched with differentially expressed genes (DEGs) were revealed. By using the molecular complex detection (MCODE) algorithm, we identified five significant clusters in the cyan module protein–protein interaction (PPI) network and nine significant clusters in the magenta module PPI network. Our analyses indicated that (i) human accelerated region (HAR) genes are enriched in the magenta-associated C6 cluster, and (ii) positive selection (PS) genes are enriched in the cyan-associated C3 and C5 clusters. We propose these enriched HAR and PS genes, i.e., EIF4E, EIF5, EIF3M, DDX17, SRSF11, PSPC1, SUMO1, CAPZA1, PSMD14, and MNAT1, including highly connected hub genes, HNRNPA1, RBMX, PRKDC, and RANBP2, as candidate key genes for PE’s pathogenesis. A further clarification of the functions of these PPI clusters and key enriched genes will contribute to the discovery of diagnostic biomarkers for PE and therapeutic intervention targets.
Collapse
Affiliation(s)
- Kuniyo Kondoh
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1, Yanagido, Gifu-City 501-1193, Gifu, Japan
- School of Nursing, Gifu University of Health Sciences, 2-92, Higashiuzura, Gifu-City 500-8281, Gifu, Japan
| | - Hiromichi Akahori
- Department of Functional Bioscience, Gifu University School of Medicine, 1-1, Yanagido, Gifu-City 501-1193, Gifu, Japan
| | - Yoshinori Muto
- Institute for Glyco-Core Research (iGCORE), Gifu University, 1-1 Yanagido, Gifu-City 501-1193, Gifu, Japan
| | - Tomoyoshi Terada
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1, Yanagido, Gifu-City 501-1193, Gifu, Japan
- Department of Functional Bioscience, Gifu University School of Medicine, 1-1, Yanagido, Gifu-City 501-1193, Gifu, Japan
- Correspondence: ; Tel.: +81-58-293-3241
| |
Collapse
|
14
|
Tarca AL, Romero R, Bhatti G, Gotsch F, Done B, Gudicha DW, Gallo DM, Jung E, Pique-Regi R, Berry SM, Chaiworapongsa T, Gomez-Lopez N. Human Plasma Proteome During Normal Pregnancy. J Proteome Res 2022; 21:2687-2702. [PMID: 36154181 PMCID: PMC10445406 DOI: 10.1021/acs.jproteome.2c00391] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The human plasma proteome is underexplored despite its potential value for monitoring health and disease. Herein, using a recently developed aptamer-based platform, we profiled 7288 proteins in 528 plasma samples from 91 normal pregnancies (Gene Expression Omnibus identifier GSE206454). The coefficient of variation was <20% for 93% of analytes (median 7%), and a cross-platform correlation for selected key angiogenic and anti-angiogenic proteins was significant. Gestational age was associated with changes in 953 proteins, including highly modulated placenta- and decidua-specific proteins, and they were enriched in biological processes including regulation of growth, angiogenesis, immunity, and inflammation. The abundance of proteins corresponding to RNAs specific to populations of cells previously described by single-cell RNA-Seq analysis of the placenta was highly modulated throughout gestation. Furthermore, machine learning-based prediction of gestational age and of time from sampling to term delivery compared favorably with transcriptomic models (mean absolute error of 2 weeks). These results suggested that the plasma proteome may provide a non-invasive readout of placental cellular dynamics and serve as a blueprint for investigating obstetrical disease.
Collapse
Affiliation(s)
- Adi L Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan48202, United States
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan48103, United States
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan48824, United States
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan48202, United States
- Detroit Medical Center, Detroit, Michigan48201, United States
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
| | - Francesca Gotsch
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
| | - Bogdan Done
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
| | - Dereje W Gudicha
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
| | - Dahiana M Gallo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, University of Valle 13, Cali, Valle del Cauca100-00, Colombia
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
| | - Roger Pique-Regi
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan48202, United States
| | - Stanley M Berry
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
| |
Collapse
|
15
|
Sharma AK, Singh S, Singh H, Mahajan D, Kolli P, Mandadapu G, Kumar B, Kumar D, Kumar S, Jena MK. Deep Insight of the Pathophysiology of Gestational Diabetes Mellitus. Cells 2022; 11:2672. [PMID: 36078079 PMCID: PMC9455072 DOI: 10.3390/cells11172672] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 12/19/2022] Open
Abstract
Diabetes mellitus is a severe metabolic disorder, which consistently requires medical care and self-management to restrict complications, such as obesity, kidney damage and cardiovascular diseases. The subtype gestational diabetes mellitus (GDM) occurs during pregnancy, which severely affects both the mother and the growing foetus. Obesity, uncontrolled weight gain and advanced gestational age are the prominent risk factors for GDM, which lead to high rate of perinatal mortality and morbidity. In-depth understanding of the molecular mechanism involved in GDM will help researchers to design drugs for the optimal management of the condition without affecting the mother and foetus. This review article is focused on the molecular mechanism involved in the pathophysiology of GDM and the probable biomarkers, which can be helpful for the early diagnosis of the condition. The early diagnosis of the metabolic disorder, most preferably in first trimester of pregnancy, will lead to its effective long-term management, reducing foetal developmental complications and mortality along with safety measures for the mother.
Collapse
Affiliation(s)
- Amarish Kumar Sharma
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Sanjeev Singh
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Himanshu Singh
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Deviyani Mahajan
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Prachetha Kolli
- Microgen Health Inc., 14225 Sullyfield Cir Suite E, Chantilly, VA 20151, USA
| | | | - Bimlesh Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Dharmendra Kumar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar 125001, Haryana, India
| | - Sudarshan Kumar
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Manoj Kumar Jena
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| |
Collapse
|
16
|
Liu Y, Zhang Q, Gao X, Wang T. Study on lipid nanomicelles targeting placenta for the treatment of preeclampsia. J Drug Target 2022; 30:894-909. [PMID: 35502921 DOI: 10.1080/1061186x.2022.2068558] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
In view of the serious clinical harm of preeclampsia and the lack of effective treatment methods, a PEG-modified lipid hybrid micelle was designed with folic acid molecule on the surface, containing siRNA, targeted delivery to the placenta, interfering the expression of sFlt-1 and treating preeclampsia. In this paper, the preparation and characterization of lipid hybrid micelles were investigated in detail, the cytology in vitro and in vivo distribution, pharmacodynamics, safety and action mechanism of the preparation were studied, which laid a foundation for gene therapy of preeclampsia.
Collapse
Affiliation(s)
- Yang Liu
- School of Pharmaceutical Sciences, Zhengzhou University, No.100, Kexue Avenue, Zhengzhou 450001, China
| | - Qimeng Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, No.100, Kexue Avenue, Zhengzhou 450001, China
| | - Xingli Gao
- School of Pharmaceutical Sciences, Zhengzhou University, No.100, Kexue Avenue, Zhengzhou 450001, China
| | - Tong Wang
- School of Pharmaceutical Sciences, Zhengzhou University, No.100, Kexue Avenue, Zhengzhou 450001, China
| |
Collapse
|
17
|
MicroRNA-495 suppresses pre-eclampsia via activation of p53/PUMA axis. Cell Death Dis 2022; 8:132. [PMID: 35338123 PMCID: PMC8956677 DOI: 10.1038/s41420-022-00874-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 01/20/2022] [Accepted: 02/08/2022] [Indexed: 11/29/2022]
Abstract
Linkage between microRNAs (miRNAs) and pre-eclampsia (PE) has been documented. Here, we focused on miR-495 in PE and its underlying mechanism in regulation of trophoblast cells. Expression of miR-495, HDAC2, p53 and PUMA was determined in collected placental tissue samples. Loss- and gain-function was performed to determine the roles of miR-495, HDAC2, p53, and PUMA in biological processes of HTR8/SVneo cells and primary trophoblast cells. The relationships among miR-495, HDAC2, and p53 were pinpointed. PE patients presented with higher expression of miR-495, p53, and PUMA in placental tissues, but lower HDAC2. miR-495 negatively targeted HDAC2 expression. HDAC2 suppressed p53 expression via deacetylation. Overexpression of miR-495, p53, or PUMA inhibited biological properties of HTR8/SVneo cells and primary trophoblast cells, while opposite trends were observed in response to oe-HDAC2. In conclusion, miR-495 knockdown can suppress p53/PUMA axis by targeting HDAC2 to enhance biological behaviors of trophoblast cells, which may prevent occurrence of PE.
Collapse
|
18
|
Low-dose aspirin prevents LPS-induced preeclampsia-like phenotype via AQP-1 and the MAPK/ERK 1/2 pathway. Placenta 2022; 121:61-69. [DOI: 10.1016/j.placenta.2022.03.007] [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: 12/07/2021] [Revised: 02/23/2022] [Accepted: 03/03/2022] [Indexed: 11/16/2022]
|
19
|
Liu L, Wang R, Xu R, Chu Y, Gu W. Procyanidin B2 ameliorates endothelial dysfunction and impaired angiogenesis via the Nrf2/PPARγ/sFlt-1 axis in preeclampsia. Pharmacol Res 2022; 177:106127. [PMID: 35150862 DOI: 10.1016/j.phrs.2022.106127] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/30/2022] [Accepted: 02/08/2022] [Indexed: 12/14/2022]
Abstract
Preeclampsia is a severe complication of pregnancy characterized by variable degrees of placental malperfusion. A growing body of evidence indicates that soluble endoglin and soluble fms-like tyrosine kinase-1 (sFlt-1) play important pathophysiological roles in preeclampsia, causing endothelial dysfunction, hypertension, and multiorgan injury. A drug that is safe in pregnancy and inhibits placental sFlt-1 and soluble endoglin secretion would be an attractive treatment strategy for preeclampsia. Procyanidin B2, a bioactive food compound, has been reported to exert multiple beneficial functions. Placental explant cultures in vitro are useful for studying tissue functions including release of secretory components, pharmacology, toxicology, and disease processes. The reduced uterine perfusion pressure (RUPP) rat model has been widely used as a model of preeclampsia. We aimed to investigate the effect of procyanidin B2 on preeclampsia via using placental explant cultures and RUPP rat model. In this study, we demonstrated that procyanidin B2 reduced soluble endoglin and sFlt-1 secretion from human umbilical vein endothelial cells (HUVECs), primary trophoblasts, and placental explants from preeclamptic pregnancies. Moreover, procyanidin B2 alleviated endothelial dysfunction and impaired angiogenesis induced by sFlt-1, including increasing the migration, invasion and angiogenesis of endothelial cells and decreasing the expression of vascular cell adhesion molecule-1 (VCAM-1) and leukocyte adhesion on HUVECs. In addition, procyanidin B2 promoted nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear accumulation and induced peroxisome proliferator-activated receptor γ (PPARγ) expression in primary placental tissues and endothelial cells. Importantly, Nrf2 specifically binds to the PPARγ promoter region (-1227/-1217) and enhances its transcriptional activity. Procyanidin B2 inhibits sFlt-1 secretion via the Nrf2/PPARγ axis. In the RUPP rat model of preeclampsia, procyanidin B2 attenuated RUPP-induced maternal angiogenic imbalance, hypertension and improved placental and fetal weight. Taken together, our results demonstrate that procyanidin B2 inhibits sFlt-1 secretion and ameliorates endothelial dysfunction and impaired angiogenesis via the Nrf2/PPARγ axis in preeclampsia. Procyanidin B2 may be a novel therapeutic agent for treatment of preeclampsia.
Collapse
Affiliation(s)
- Lei Liu
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Rencheng Wang
- Department of Obstetrics and Gynecology, Renhe Hospital Baoshan District, Shanghai 200431, China
| | - Ran Xu
- Department of Obstetrics and Gynecology, Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, 310030, China
| | - Yuening Chu
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Weirong Gu
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China.
| |
Collapse
|
20
|
Preeclampsia and eclampsia: the conceptual evolution of a syndrome. Am J Obstet Gynecol 2022; 226:S786-S803. [PMID: 35177220 PMCID: PMC8941666 DOI: 10.1016/j.ajog.2021.12.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 02/03/2023]
Abstract
Preeclampsia, one of the most enigmatic complications of pregnancy, is considered a pregnancy-specific disorder caused by the placenta and cured only by delivery. This article traces the condition from its origins-once thought to be a disease of the central nervous system, recognized by the occurrence of seizures (ie, eclampsia)-to the present time when preeclampsia is conceptualized primarily as a vascular disorder. We review the epidemiologic data that led to the recommendation to use diastolic hypertension and proteinuria as diagnostic criteria, as their combined presence was associated with an increased risk of fetal death and the birth of small-for-gestational-age neonates. However, preeclampsia is a multisystemic disorder with protean manifestations, and the condition can be present even in the absence of hypertension and proteinuria. Toxins gaining access to the maternal circulation have been proposed to mediate the clinical manifestations-hence, the term "toxemia of pregnancy," which was used for several decades. The search for putative toxins has challenged investigators for more than a century, and a growing body of evidence suggests that products of an ischemic or a stressed placenta are responsible for the vascular changes that characterize this syndrome. The discovery that the placenta can produce antiangiogenic factors, which regulate endothelial cell function and induce intravascular inflammation, has been a major step forward in the understanding of preeclampsia. We view the release of antiangiogenic factors by the placenta as an adaptive response to improve uterine perfusion by modulating endothelial function and maternal cardiovascular performance. However, this homeostatic response can become maladaptive and lead to damage of target organs during pregnancy or the postpartum period. Early-onset preeclampsia has many features in common with atherosclerosis, whereas late-onset preeclampsia seems to result from a mismatch of fetal demands and maternal supply, that is, a metabolic crisis. Preeclampsia, as it is understood today, is essentially vascular dysfunction unmasked or caused by pregnancy. A subset of patients diagnosed with preeclampsia are at greater risk of the subsequent development of hypertension, ischemic heart disease, heart failure, vascular dementia, and end-stage renal disease. However, these adverse events may be the result of a preexisting vascular pathologic process; it is not known if the occurrence of preeclampsia increases the baseline risk. Therefore, the understanding, prediction, prevention, and treatment of preeclampsia are healthcare priorities.
Collapse
|
21
|
Gusar V, Timofeeva A, Chagovets V, Kan N, Vysokikh M, Marey M, Karapetyan A, Baev O, Sukhikh G. Diagnostic Potential of Exosomal HypoxamiRs in the Context of Hypoxia-Sumoylation-HypoxamiRs in Early Onset Preeclampsia at the Preclinical Stage. LIFE (BASEL, SWITZERLAND) 2022; 12:life12010101. [PMID: 35054494 PMCID: PMC8780366 DOI: 10.3390/life12010101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/04/2022] [Accepted: 01/10/2022] [Indexed: 11/16/2022]
Abstract
As the search for non-invasive preclinical markers of preeclampsia (PE) expands, the number of studies on the diagnostic potential of exosomes is growing. Changes in the partial pressure of oxygen caused by impaired uteroplacental perfusion in PE are a powerful inducer of increased production and release of exosomes from cells, which also determine their cargo. At the same time, the expression pattern of oxygen-dependent microRNAs (miRNAs), called "hypoxamiRs", is modulated, and their packing into exosomes is strictly regulated by sumoylation. In connection therewith, we emphasize the evaluation of exosomal hypoxamiR expression (miR-27b-3p, miR-92b-3p, miR-181a-5p, and miR-186-5p) using quantitative RT-PCR, as well as SUMO 1-4 and UBC9 (by Western blotting), in pregnant women with early-onset PE. The findings show that miR-27b-3p and miR-92b-3p expression was significantly changed at 11-14 and 24-26 weeks of gestation in the blood plasma of pregnant women with early-onset PE, which subsequently manifested. High sensitivity and specificity (AUC = 1) were demonstrated for these miRNAs in the first trimester, and significant correlations with a decrease in hemoglobin (r = 0.71, p = 0.002; r = -0.71, p = 0.002) were established. In mid-pregnancy, the miR-27b-3p expression was found to correlate with an increase in platelets (r = -0.95, p = 0.003), and miR-92b-3p was associated with a decrease in the prothrombin index (r = 0.95, p = 0.003). Specific exomotifs of studied miRNAs were also identified, to which the sumoylated ribonucleoprotein hnRNPA2/B1 binds, carrying out their packaging into exosomes. The expression of conjugated SUMO 1 (p = 0.05), SUMO 2/3/4 (p = 0.03), and UBC9 (p = 0.1) was increased in exosomes at early-onset PE, and the expression of free SUMO 1 (p = 0.03) and SUMO 2/3/4 (p = 0.01) was significantly increased in the placenta, as an adaptive response to hypoxia. Moreover, SUMO 2/3/4 was negatively correlated with miR-27b-3p expression in the placenta. In conclusion, the diagnostic potential of exosomal hypoxamiRs mediated by sumoylation may form the basis for the development of combined specific targets for the treatment of early-onset PE, as hnRNPA2/B1 is a target of miR-27b-3p, and its sumoylation creates miR-27b-3p-hnRNPA2/B1-SUMO 1-4 cross-talk.
Collapse
Affiliation(s)
- Vladislava Gusar
- Laboratory of Applied Transcriptomics, Federal State Budget Institution, “National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov” of the Ministry of Healthcare of the Russian Federation, Oparin Str. 4, 117997 Moscow, Russia;
- Correspondence:
| | - Angelika Timofeeva
- Laboratory of Applied Transcriptomics, Federal State Budget Institution, “National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov” of the Ministry of Healthcare of the Russian Federation, Oparin Str. 4, 117997 Moscow, Russia;
| | - Vitaliy Chagovets
- Laboratory of Proteomics and Metabolomics of Human Reproduction, Federal State Budget Institution, “National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov” of the Ministry of Healthcare of the Russian Federation, Oparin Str. 4, 117997 Moscow, Russia;
| | - Nataliya Kan
- Directorat, Federal State Budget Institution, “National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov” of the Ministry of Healthcare of the Russian Federation, Oparin Str. 4, 117997 Moscow, Russia; (N.K.); (G.S.)
| | - Mikhail Vysokikh
- Laboratory of Mitochondrial Medicine, Federal State Budget Institution, “National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov” of the Ministry of Healthcare of the Russian Federation, Oparin Str. 4, 117997 Moscow, Russia; (M.V.); (M.M.)
| | - Maria Marey
- Laboratory of Mitochondrial Medicine, Federal State Budget Institution, “National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov” of the Ministry of Healthcare of the Russian Federation, Oparin Str. 4, 117997 Moscow, Russia; (M.V.); (M.M.)
| | - Anna Karapetyan
- Maternity Department, Federal State Budget Institution, “National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov” of the Ministry of Healthcare of the Russian Federation, Oparin Str. 4, 117997 Moscow, Russia; (A.K.); (O.B.)
| | - Oleg Baev
- Maternity Department, Federal State Budget Institution, “National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov” of the Ministry of Healthcare of the Russian Federation, Oparin Str. 4, 117997 Moscow, Russia; (A.K.); (O.B.)
| | - Gennadiy Sukhikh
- Directorat, Federal State Budget Institution, “National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov” of the Ministry of Healthcare of the Russian Federation, Oparin Str. 4, 117997 Moscow, Russia; (N.K.); (G.S.)
| |
Collapse
|
22
|
First Trimester Mean Platelet Volume, Neutrophil to Lymphocyte Ratio, and Platelet to Lymphocyte Ratio Values Are Useful Markers for Predicting Preeclampsia. Ochsner J 2022; 21:364-370. [PMID: 34984051 PMCID: PMC8675624 DOI: 10.31486/toj.21.0026] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Background: Preeclampsia complicates 2% to 8% of all pregnancies. Systemic inflammatory response (SIR) markers are widely used in the diagnosis of many inflammatory diseases and in the prediction of complicated pregnancies. This study examined the diagnostic value of SIR markers during the first trimester of pregnancy to predict preeclampsia development. Methods: This retrospective case-control study was conducted from January 2020 to May 2020. We included 94 patients diagnosed with mild preeclampsia, 107 patients diagnosed with severe preeclampsia, and 100 normotensive pregnant patients as controls. We obtained the first trimester (6 to 14 weeks) complete blood cell counts for all patients. We used a receiver operating characteristic curve to evaluate the cutoff, sensitivity, and specificity values. Results: First trimester mean platelet volume (MPV), neutrophil to lymphocyte ratio (NLR), and platelet to lymphocyte ratio (PLR) values were significantly higher in patients who developed preeclampsia in later pregnancy weeks. The optimal cutoff value for MPV was 10.65 fL, with a sensitivity of 63.7% and a specificity of 65.0%. The best predictor for preeclampsia was NLR at an optimal cutoff value of 4.12, with a sensitivity of 82.1% and specificity of 62.0%. At a cutoff value of 131.8, PLR predicted preeclampsia with a sensitivity rate of 65.0% and a specificity rate of 60.2%. Conclusion: The results of this study suggest that first trimester MPV, NLR, and PLR values are clinically useful markers in the prediction of preeclampsia. The increased first trimester values of MPV, NLR, and PLR also indicate that inflammation may play a crucial role in preeclampsia pathogenesis.
Collapse
|
23
|
Prediction of preeclampsia throughout gestation with maternal characteristics and biophysical and biochemical markers: a longitudinal study. Am J Obstet Gynecol 2022; 226:126.e1-126.e22. [PMID: 34998477 PMCID: PMC8749051 DOI: 10.1016/j.ajog.2021.01.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/22/2021] [Accepted: 01/22/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND The current approach to predict preeclampsia combines maternal risk factors and evidence from biophysical markers (mean arterial pressure, Doppler velocimetry of the uterine arteries) and maternal blood proteins (placental growth factor, soluble vascular endothelial growth factor receptor-1, pregnancy-associated plasma protein A). Such models require the transformation of biomarker data into multiples of the mean values by using population- and site-specific models. Previous studies have focused on a narrow window in gestation and have not included the maternal blood concentration of soluble endoglin, an important antiangiogenic factor up-regulated in preeclampsia. OBJECTIVE This study aimed (1) to develop models for the calculation of multiples of the mean values for mean arterial pressure and biochemical markers; (2) to build and assess the predictive models for preeclampsia based on maternal risk factors, the biophysical (mean arterial pressure) and biochemical (placental growth factor, soluble vascular endothelial growth factor receptor-1, and soluble endoglin) markers collected throughout pregnancy; and (3) to evaluate how prediction accuracy is affected by the presence of chronic hypertension and gestational age. STUDY DESIGN This longitudinal case-cohort study included 1150 pregnant women: women without preeclampsia with (n=49) and without chronic hypertension (n=871) and those who developed preeclampsia (n=166) or superimposed preeclampsia (n=64). Mean arterial pressure and immunoassay-based maternal plasma placental growth factor, soluble vascular endothelial growth factor receptor-1, and soluble endoglin concentrations were available throughout pregnancy (median of 5 observations per patient). A prior-risk model for preeclampsia was established by using Poisson regression based on maternal characteristics and obstetrical history. Next, multiple regression was used to fit biophysical and biochemical marker data as a function of maternal characteristics by using data collected at 8 to 15+6, 16 to 19+6, 20 to 23+6, 24 to 27+6, 28 to 31+6, and 32 to 36+6 week intervals, and observed values were converted into multiples of the mean values. Then, multivariable prediction models for preeclampsia were fit based on the biomarker multiples of the mean data and prior-risk estimates. Separate models were derived for overall, preterm, and term preeclampsia, which were evaluated by receiver operating characteristic curves and sensitivity at fixed false-positive rates. RESULTS (1) The inclusion of soluble endoglin in prediction models for all preeclampsia, together with the prior-risk estimates, mean arterial pressure, placental growth factor, and soluble vascular endothelial growth factor receptor-1, increased the sensitivity (at a fixed false-positive rate of 10%) for early prediction of superimposed preeclampsia, with the largest increase (from 44% to 54%) noted at 20 to 23+6 weeks (McNemar test, P<.05); (2) combined evidence from prior-risk estimates and biomarkers predicted preterm preeclampsia with a sensitivity (false-positive rate, 10%) of 55%, 48%, 62%, 72%, and 84% at 8 to 15+6, 16 to 19+6, 20 to 23+6, 24 to 27+6, and 28 to 31+6 week intervals, respectively; (3) the sensitivity for term preeclampsia (false-positive rate, 10%) was 36%, 36%, 41%, 43%, 39%, and 51% at 8 to 15+6, 16 to 19+6, 20 to 23+6, 24 to 27+6, 28 to 31+6, and 32 to 36+6 week intervals, respectively; (4) the detection rate for superimposed preeclampsia among women with chronic hypertension was similar to that in women without chronic hypertension, especially earlier in pregnancy, reaching at most 54% at 20 to 23+6 weeks (false-positive rate, 10%); and (5) prediction models performed comparably to the Fetal Medicine Foundation calculators when the same maternal risk factors and biomarkers (mean arterial pressure, placental growth factor, and soluble vascular endothelial growth factor receptor-1 multiples of the mean values) were used as input. CONCLUSION We introduced prediction models for preeclampsia throughout pregnancy. These models can be useful to identify women at risk during the first trimester who could benefit from aspirin treatment or later in pregnancy to inform patient management. Relative to prediction performance at 8 to 15+6 weeks, there was a substantial improvement in the detection rate for preterm and term preeclampsia by using data collected after 20 and 32 weeks' gestation, respectively. The inclusion of plasma soluble endoglin improves the early prediction of superimposed preeclampsia, which may be valuable when Doppler velocimetry of the uterine arteries is not available.
Collapse
|
24
|
Huang Q, Hao S, You J, Yao X, Li Z, Schilling J, Thyparambil S, Liao WL, Zhou X, Mo L, Ladella S, Davies-Balch SR, Zhao H, Fan D, Whitin JC, Cohen HJ, McElhinney DB, Wong RJ, Shaw GM, Stevenson DK, Sylvester KG, Ling XB. Early-pregnancy prediction of risk for pre-eclampsia using maternal blood leptin/ceramide ratio: discovery and confirmation. BMJ Open 2021; 11:e050963. [PMID: 34824115 PMCID: PMC8627403 DOI: 10.1136/bmjopen-2021-050963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE This study aimed to develop a blood test for the prediction of pre-eclampsia (PE) early in gestation. We hypothesised that the longitudinal measurements of circulating adipokines and sphingolipids in maternal serum over the course of pregnancy could identify novel prognostic biomarkers that are predictive of impending event of PE early in gestation. STUDY DESIGN Retrospective discovery and longitudinal confirmation. SETTING Maternity units from two US hospitals. PARTICIPANTS Six previously published studies of placental tissue (78 PE and 95 non-PE) were compiled for genomic discovery, maternal sera from 15 women (7 non-PE and 8 PE) enrolled at ProMedDx were used for sphingolipidomic discovery, and maternal sera from 40 women (20 non-PE and 20 PE) enrolled at Stanford University were used for longitudinal observation. OUTCOME MEASURES Biomarker candidates from discovery were longitudinally confirmed and compared in parallel to the ratio of placental growth factor (PlGF) and soluble fms-like tyrosine kinase (sFlt-1) using the same cohort. The datasets were generated by enzyme-linked immunosorbent and liquid chromatography-tandem mass spectrometric assays. RESULTS Our discovery integrating genomic and sphingolipidomic analysis identified leptin (Lep) and ceramide (Cer) (d18:1/25:0) as novel biomarkers for early gestational assessment of PE. Our longitudinal observation revealed a marked elevation of Lep/Cer (d18:1/25:0) ratio in maternal serum at a median of 23 weeks' gestation among women with impending PE as compared with women with uncomplicated pregnancy. The Lep/Cer (d18:1/25:0) ratio significantly outperformed the established sFlt-1/PlGF ratio in predicting impending event of PE with superior sensitivity (85% vs 20%) and area under curve (0.92 vs 0.52) from 5 to 25 weeks of gestation. CONCLUSIONS Our study demonstrated the longitudinal measurement of maternal Lep/Cer (d18:1/25:0) ratio allows the non-invasive assessment of PE to identify pregnancy at high risk in early gestation, outperforming the established sFlt-1/PlGF ratio test.
Collapse
Affiliation(s)
| | - Shiying Hao
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California, USA
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Palo Alto, California, USA
| | - Jin You
- Department of Bioengineering, University of California Riverside, Riverside, California, USA
| | | | - Zhen Li
- Department of Surgery, Stanford University, Stanford, California, USA
- Binhai Industrial Technology Research Institute, Zhejiang University, Tianjin, China
- School of Electrical Engineering, Southeast University, Nanjing, China
| | | | | | | | - Xin Zhou
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China
| | - Lihong Mo
- Department of Obstetrics and Gynecology, University of California San Francisco, Fresno, California, USA
| | - Subhashini Ladella
- Department of Obstetrics and Gynecology, University of California San Francisco, Fresno, California, USA
| | | | - Hangyi Zhao
- Department of Mathematics, Stanford University, Stanford, California, USA
| | - David Fan
- Department of Statistics and Applied Probability, University of California Santa Barbara, Santa Barbara, California, USA
| | - John C Whitin
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Harvey J Cohen
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Doff B McElhinney
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California, USA
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Palo Alto, California, USA
| | - Ronald J Wong
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Gary M Shaw
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - David K Stevenson
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Karl G Sylvester
- Department of Surgery, Stanford University, Stanford, California, USA
| | - Xuefeng B Ling
- Department of Surgery, Stanford University, Stanford, California, USA
| |
Collapse
|
25
|
Rosario FJ, Pardo S, Michelsen TM, Erickson K, Moore L, Powell TL, Weintraub ST, Jansson T. Characterization of the Primary Human Trophoblast Cell Secretome Using Stable Isotope Labeling With Amino Acids in Cell Culture. Front Cell Dev Biol 2021; 9:704781. [PMID: 34595166 PMCID: PMC8476785 DOI: 10.3389/fcell.2021.704781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/16/2021] [Indexed: 12/29/2022] Open
Abstract
The placental villus syncytiotrophoblast, the nutrient-transporting and hormone-producing epithelium of the human placenta, is a critical regulator of fetal development and maternal physiology. However, the identities of the proteins synthesized and secreted by primary human trophoblast (PHT) cells remain unknown. Stable Isotope Labeling with Amino Acids in Cell Culture followed by mass spectrometry analysis of the conditioned media was used to identify secreted proteins and obtain information about their relative rates of synthesis in syncytialized multinucleated PHT cells isolated from normal term placental villus tissue (n = 4/independent placenta). A total of 1,344 proteins were identified, most of which have not previously been reported to be secreted by the human placenta or trophoblast. The majority of secreted proteins are involved in energy and carbon metabolism, glycolysis, biosynthesis of amino acids, purine metabolism, and fatty acid degradation. Histone family proteins and mitochondrial proteins were among proteins with the slowest synthesis rate whereas proteins associated with signaling and the plasma membrane were synthesized rapidly. There was a significant overlap between the PHT secretome and proteins known be secreted to the fetal circulation by the human placenta in vivo. The generated data will guide future experiments to determine the function of individual secreted proteins and will help us better understand how the placenta controls maternal and fetal physiology.
Collapse
Affiliation(s)
- Fredrick J Rosario
- Division of Reproductive Sciences, Department of OB/GYN, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Sammy Pardo
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Trond M Michelsen
- Division of Obstetrics and Gynecology, Department of Obstetrics Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Kathryn Erickson
- Division of Reproductive Sciences, Department of OB/GYN, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Lorna Moore
- Division of Reproductive Sciences, Department of OB/GYN, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Theresa L Powell
- Division of Reproductive Sciences, Department of OB/GYN, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Susan T Weintraub
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of OB/GYN, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
26
|
Phan K, Schiller I, Dendukuri N, Gomez YH, Gorgui J, El-Messidi A, Gagnon R, Daskalopoulou SS. A longitudinal analysis of arterial stiffness and wave reflection in preeclampsia: Identification of changepoints. Metabolism 2021; 120:154794. [PMID: 33971204 DOI: 10.1016/j.metabol.2021.154794] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/27/2021] [Accepted: 05/01/2021] [Indexed: 11/19/2022]
Abstract
PURPOSE Preeclampsia (PrE) is a leading complication of pregnancy characterized by vascular dysfunction. Characterizing the longitudinal changes in vascular function prior to PrE onset is critical to the identification of optimal timepoints for vascular assessment and the development of effective early screening strategies. METHODS In this prospective longitudinal study of women with singleton high-risk pregnancies, arterial stiffness and wave reflection parameters were assessed using applanation tonometry at 10-13 weeks' gestation and repeated every 4 weeks throughout pregnancy. Changepoints in carotid-femoral pulse wave velocity (cfPWV), carotid-radial PWV (crPWV), augmentation index (AIx), time to wave reflection (T1R), pulse pressure amplification (PPA), and subendocardial viability ratio (SEVR) were compared between women who did and did not subsequently develop PrE. RESULTS A changepoint in cfPWV and crPWV was detected at 14-17 weeks' gestation. cfPWV then increased in women who went on to develop PrE but decreased in women who did not; a 1.2 m/s difference in cfPWV between the groups was observed at 22-25 weeks' gestation. Conversely, crPWV converged in the two groups from a baseline difference of 1.05 m/s (95% credible interval: 0.37, 1.72). Women who subsequently developed PrE demonstrated an increase in AIx at 18-21 weeks' gestation that was not seen in women who did not develop PrE until 30-33 weeks. No differences in T1R, PPA, or SEVR were observed between the groups. CONCLUSIONS Altered vascular adaptations were detected using measures of arterial stiffness and wave reflection in the early second trimester of pregnant women who developed PrE compared to those who did not. These findings demonstrate the potential clinical utility of arterial stiffness and wave reflection parameters as an early screening tool for PrE, which can be used to inform clinical management of high-risk pregnancies.
Collapse
Affiliation(s)
- Kim Phan
- Division of Experimental Medicine, Department of Medicine, McGill University, 1001 Décarie Boulevard, Montreal, Quebec H4A 3J1, Canada.
| | - Ian Schiller
- Division of Clinical Epidemiology, McGill University Health Centre - Research Institute, 1001 Décarie Boulevard, Montreal, Quebec H4A 3J1, Canada.
| | - Nandini Dendukuri
- Division of Clinical Epidemiology, McGill University Health Centre - Research Institute, 1001 Décarie Boulevard, Montreal, Quebec H4A 3J1, Canada.
| | - Yessica-Haydee Gomez
- Division of Internal Medicine, Department of Medicine, McGill University, 1001 Décarie Boulevard, Montreal, Quebec H4A 3J1, Canada
| | - Jessica Gorgui
- Division of Internal Medicine, Department of Medicine, McGill University, 1001 Décarie Boulevard, Montreal, Quebec H4A 3J1, Canada
| | - Amira El-Messidi
- Department of Obstetrics and Gynecology, McGill University, 1001 Décarie Boulevard, Montreal, Quebec H4A 3J1, Canada.
| | - Robert Gagnon
- Department of Obstetrics and Gynecology, McGill University, 1001 Décarie Boulevard, Montreal, Quebec H4A 3J1, Canada.
| | - Stella S Daskalopoulou
- Division of Experimental Medicine, Department of Medicine, McGill University, 1001 Décarie Boulevard, Montreal, Quebec H4A 3J1, Canada; Division of Internal Medicine, Department of Medicine, McGill University, 1001 Décarie Boulevard, Montreal, Quebec H4A 3J1, Canada.
| |
Collapse
|
27
|
Komaroff AL, Rizzo R, Ecker JL. Human Herpesviruses 6A and 6B in Reproductive Diseases. Front Immunol 2021; 12:648945. [PMID: 33841432 PMCID: PMC8027340 DOI: 10.3389/fimmu.2021.648945] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/09/2021] [Indexed: 11/13/2022] Open
Abstract
Human herpesviruses 6A (HHV-6A) and human herpesvirus 6B (HHV-6B)—collectively, HHV-6A/B—are recently-discovered but ancient human viruses. The vast majority of people acquire one or both viruses, typically very early in life, producing an ineradicable lifelong infection. The viruses have been linked to several neurological, pulmonary and hematological diseases. In early human history, the viruses on multiple occasions infected a germ cell, and integrated their DNA into a human chromosome. As a result, about 1% of humans are born with the full viral genome present in every cell, with uncertain consequences for health. HHV-6A may play a role in 43% of cases of primary unexplained infertility. Both the inherited and acquired viruses may occasionally trigger several of the factors that are important in the pathogenesis of preeclampsia. Transplacental infection occurs in 1-2% of pregnancies, with some evidence suggesting adverse health consequences for the child. While emerging knowledge about these viruses in reproductive diseases is not sufficient to suggest any changes in current practice, we write this review to indicate the need for further research that could prove practice-changing.
Collapse
Affiliation(s)
- Anthony L Komaroff
- Division of General Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Roberta Rizzo
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy
| | - Jeffrey L Ecker
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
28
|
Ghaemi MS, Tarca AL, Romero R, Stanley N, Fallahzadeh R, Tanada A, Culos A, Ando K, Han X, Blumenfeld YJ, Druzin ML, El-Sayed YY, Gibbs RS, Winn VD, Contrepois K, Ling XB, Wong RJ, Shaw GM, Stevenson DK, Gaudilliere B, Aghaeepour N, Angst MS. Proteomic signatures predict preeclampsia in individual cohorts but not across cohorts - implications for clinical biomarker studies. J Matern Fetal Neonatal Med 2021; 35:5621-5628. [PMID: 33653202 PMCID: PMC8410912 DOI: 10.1080/14767058.2021.1888915] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Background: Early identification of pregnant women at risk for preeclampsia (PE) is important, as it will enable targeted interventions ahead of clinical manifestations. The quantitative analyses of plasma proteins feature prominently among molecular approaches used for risk prediction. However, derivation of protein signatures of sufficient predictive power has been challenging. The recent availability of platforms simultaneously assessing over 1000 plasma proteins offers broad examinations of the plasma proteome, which may enable the extraction of proteomic signatures with improved prognostic performance in prenatal care. Objective: The primary aim of this study was to examine the generalizability of proteomic signatures predictive of PE in two cohorts of pregnant women whose plasma proteome was interrogated with the same highly multiplexed platform. Establishing generalizability, or lack thereof, is critical to devise strategies facilitating the development of clinically useful predictive tests. A second aim was to examine the generalizability of protein signatures predictive of gestational age (GA) in uncomplicated pregnancies in the same cohorts to contrast physiological and pathological pregnancy outcomes. Study design: Serial blood samples were collected during the first, second, and third trimesters in 18 women who developed PE and 18 women with uncomplicated pregnancies (Stanford cohort). The second cohort (Detroit), used for comparative analysis, consisted of 76 women with PE and 90 women with uncomplicated pregnancies. Multivariate analyses were applied to infer predictive and cohort-specific proteomic models, which were then tested in the alternate cohort. Gene ontology (GO) analysis was performed to identify biological processes that were over-represented among top-ranked proteins associated with PE. Results: The model derived in the Stanford cohort was highly significant (p = 3.9E–15) and predictive (AUC = 0.96), but failed validation in the Detroit cohort (p = 9.7E–01, AUC = 0.50). Similarly, the model derived in the Detroit cohort was highly significant (p = 1.0E–21, AUC = 0.73), but failed validation in the Stanford cohort (p = 7.3E–02, AUC = 0.60). By contrast, proteomic models predicting GA were readily validated across the Stanford (p = 1.1E–454, R = 0.92) and Detroit cohorts (p = 1.1.E–92, R = 0.92) indicating that the proteomic assay performed well enough to infer a generalizable model across studied cohorts, which makes it less likely that technical aspects of the assay, including batch effects, accounted for observed differences. Conclusions: Results point to a broader issue relevant for proteomic and other omic discovery studies in patient cohorts suffering from a clinical syndrome, such as PE, driven by heterogeneous pathophysiologies. While novel technologies including highly multiplex proteomic arrays and adapted computational algorithms allow for novel discoveries for a particular study cohort, they may not readily generalize across cohorts. A likely reason is that the prevalence of pathophysiologic processes leading up to the “same” clinical syndrome can be distributed differently in different and smaller-sized cohorts. Signatures derived in individual cohorts may simply capture different facets of the spectrum of pathophysiologic processes driving a syndrome. Our findings have important implications for the design of omic studies of a syndrome like PE. They highlight the need for performing such studies in diverse and well-phenotyped patient populations that are large enough to characterize subsets of patients with shared pathophysiologies to then derive subset-specific signatures of sufficient predictive power.
Collapse
Affiliation(s)
- Mohammad S Ghaemi
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Adi L Tarca
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Natalie Stanley
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ramin Fallahzadeh
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Athena Tanada
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Anthony Culos
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Kazuo Ando
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaoyuan Han
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Yair J Blumenfeld
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Maurice L Druzin
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yasser Y El-Sayed
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ronald S Gibbs
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kevin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Xuefeng B Ling
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ronald J Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Gary M Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - David K Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.,Department of Biomedical Data Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
29
|
von Hellens H, Keski-Nisula L, Sahlman H. Increased risk of preeclampsia after use of paracetamol during pregnancy - causal or coincidence? BMC Pregnancy Childbirth 2021; 21:24. [PMID: 33407239 PMCID: PMC7789579 DOI: 10.1186/s12884-020-03490-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 12/10/2020] [Indexed: 01/31/2023] Open
Abstract
Background The maternal use of paracetamol during pregnancy has been associated with the development of preeclampsia. This study aims to clarify whether the connection is causal or whether it is due to reverse causation. Methods This study is a continuation of the retrospective case cohort study examining 2,508 pregnant women using a variety of drugs and the development of preeclampsia (1,252 women with preeclampsia and 1,256 controls). For the purposes of this study, more precise data was collected from several hospital databases of the women among this cohort who had reported taking paracetamol during pregnancy (indications, gestational period etc.); this was evaluated in association with the development of preeclampsia. Results 5.5% (100 cases and 37 controls) of all the study population (2,508) had clearly reported paracetamol use. Women with preeclampsia had used significantly more often paracetamol during pregnancy compared to controls (cases 8.0%, controls 2.9%, p < 0.001). The difference was most evident in the third trimester (after the 29th GW) and the use of paracetamol was associated with both mild and severe preeclampsia. Headache and “general pain” were the most common indications for medication among all paracetamol users. Conclusions The use of paracetamol in the third trimester of pregnancy was associated with preeclampsia. This observation indicates that association between paracetamol use and preeclampsia is probably due to reverse causation, i.e. women with preeclampsia experience more headaches due to preeclampsia symptoms since this association was not detected with the use of paracetamol in earlier stages of pregnancy.
Collapse
Affiliation(s)
- Hetti von Hellens
- Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Leea Keski-Nisula
- Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.,Department of Obstetrics and Gynecology, Kuopio University Hospital, Puijonlaaksontie 2, 70210, Kuopio, Finland
| | - Heidi Sahlman
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
30
|
Nyuydzefon B, Bonaventure J, Raissa Bifouna I, Agnes E. Complications of severe pre-eclampsia associated with acute intestinal intussusception—A case report. JOURNAL OF OBSTETRIC ANAESTHESIA AND CRITICAL CARE 2021. [DOI: 10.4103/joacc.joacc_93_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
31
|
Zhang N, Tan J, Yang H, Khalil RA. Comparative risks and predictors of preeclamptic pregnancy in the Eastern, Western and developing world. Biochem Pharmacol 2020; 182:114247. [PMID: 32986983 PMCID: PMC7686229 DOI: 10.1016/j.bcp.2020.114247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 11/15/2022]
Abstract
Preeclampsia (PE) is a complication of pregnancy characterized by hypertension (HTN-Preg), and often proteinuria. If not managed promptly, PE could lead to eclampsia and seizures. PE could also lead to intrauterine growth restriction (IUGR) and prematurity at birth. Although PE is a major cause of maternal and fetal morbidity and mortality, the underlying mechanisms are unclear. Also, there is a wide variability in the incidence of PE, ranging between 2 and 8% of pregnancies in the Eastern, Western and Developing world, suggesting regional differences in the risk factors and predictors of the pregnancy-related disorder. Several demographic, genetic, dietary and environmental factors, as well as maternal circulating biomarkers have been associated with PE. Demographic factors such as maternal race and ethnicity could play a role in PE. Specific genetic polymorphisms have been identified in PE. Maternal age, parity, education and socioeconomic status could be involved in PE. Dietary fat, protein, calcium and vitamins, body weight, and environmental factors including climate changes and air pollutants could also play a role in PE. Several circulating cytoactive factors including anti-angiogenic factors and cytokines have also been associated with PE. Traditional midwifery care is a common practice in local maternity care units, while advanced perinatal care and new diagnostic tools such as uterine artery Doppler velocimetry have been useful in predicting early PE in major medical centers. These PE risk factors, early predictors and diagnostic tools vary vastly in different regions of the Eastern, Western and Developing world. Further understanding of the differences in the demographic, genetic, dietary and environmental factors among pregnant women in different world regions should help in designing a region-specific cluster of risk factors and predictors of PE, and in turn provide better guidance for region-specific tools for early detection and management of PE.
Collapse
Affiliation(s)
- Ning Zhang
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jing Tan
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - HaiFeng Yang
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
32
|
Transfer of miR-15a-5p by placental exosomes promotes pre-eclampsia progression by regulating PI3K/AKT signaling pathway via CDK1. Mol Immunol 2020; 128:277-286. [PMID: 33197745 DOI: 10.1016/j.molimm.2020.10.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 01/03/2023]
Abstract
Preeclampsia (PE) is a systemic complication that occurs after the 20th week of gestation and is characterized by the onset of hypertension and proteinuria. Dysregulated circulating microRNA (miRNA) has usually been noted in PE. Understanding the release profile and bioactivity of placental exosomes is a promising mode of identifying dysregulated miRNA, which may be useful biomarkers of PE. Herein, we aimed to investigate the role of placental exosomes and their miRNA cargo miR-15a-5p in PE. miR-15a-5p was found upregulated in exosomes isolated from maternal plasma of PE pregnant women as compared to those from normal pregnant women. Placental exosomes derived from PE pregnant women suppressed the proliferation and invasion of HTR-8/SVneo cells but promoted cell apoptosis, which was dictated by their cargo miR-15a-5p. Further investigation showed that exosomal miR-15a-5p inhibited the activation of the PI3K/AKT pathway by down-regulating CDK1, thus suppressing HTR-8/SVneo cell proliferation, invasion, and apoptosis. In vivo analysis demonstrated that placental exosomes treated with miR-15a-5p inhibitor attenuated histopathologic changes and apoptosis in the placenta of PE mice. In conclusion, these results provided evidence that transfer of miR-15a-5p by placental exosomes could be a promising therapeutic target to combat PE.
Collapse
|
33
|
Liu E, Zhou Y, Li J, Zhang D. MicroRNA‑491‑5p inhibits trophoblast cell migration and invasion through targeting matrix metalloproteinase‑9 in preeclampsia. Mol Med Rep 2020; 22:5033-5040. [PMID: 33174053 PMCID: PMC7646938 DOI: 10.3892/mmr.2020.11604] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/04/2020] [Indexed: 01/03/2023] Open
Abstract
Insufficient invasion of trophoblasts is correlated with the development of preeclampsia (PE). MicroRNA (miR)-491-5p has been reported to be implicated in human cancer cell invasion; however, whether miR-491-5p is involved in the development of PE remains largely unclear. The aim of the present study was to investigate the role of miR-491-5p in trophoblastic invasion in vitro and to determine its underlying mechanism of action. The expression levels of miR-491-5p were validated using reverse transcription-quantitative PCR. The effects of miR-491-5p on trophoblast cell invasion were evaluated in vitro. Then, the association between miR-491-5p and its downstream target was investigated in both cell lines and clinical specimens. miR-491-5p expression levels were observed to be significantly increased in the placental tissues from patients with PE. The invasive capacity of HTR-8/SVneo trophoblast cells was suppressed following the upregulation of miR-491-5p and increased following the inhibition of miR-491-5p. Matrix metalloproteinase-9 (MMP-9), a well-known regulator of trophoblast cell invasion, was discovered to be a direct target of miR-491-5p in HTR-8/SVneo trophoblast cells. Moreover, miR-491-5p expression levels were found to be inversely correlated with MMP-9 expression levels in placental tissues from patients with PE. The overexpression of MMP-9 partly attenuated the inhibitory effects of miR-491-5p on HTR-8/SVneo trophoblast cells invasion. Collectively, these findings suggested that the aberrant expression of miR-491-5p may contribute to PE through suppressing trophoblast invasion, thus highlighting the novel roles of miR-491-5p in the molecular pathogenesis of PE. The present study also showed that the miR-491-5p/MMP-9 axis may be an effective biomarker or a viable drug target for therapeutic intervention in PE.
Collapse
Affiliation(s)
- Enling Liu
- Department of Obstetrics and Gynecology, Tangshan Gongren Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| | - Yuxiu Zhou
- Department of Immunity, Tangshan Gongren Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| | - Jun Li
- Department of Obstetrics and Gynecology, Tangshan Gongren Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| | - Donghong Zhang
- Department of Obstetrics and Gynecology, Tangshan Gongren Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
34
|
Banala C, Moreno S, Cruz Y, Boelig RC, Saccone G, Berghella V, Schoen CN, Roman A. Impact of the ACOG guideline regarding low-dose aspirin for prevention of superimposed preeclampsia in women with chronic hypertension. Am J Obstet Gynecol 2020; 223:419.e1-419.e16. [PMID: 32173446 DOI: 10.1016/j.ajog.2020.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 02/28/2020] [Accepted: 03/03/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Patients with chronic hypertension are at increased risk for superimposed preeclampsia. The 2016 American College of Obstetricians and Gynecologists guideline recommended initiating 81 mg of daily aspirin for all pregnant women with chronic hypertension to prevent superimposed preeclampsia. OBJECTIVE (1) To evaluate the rates of implementation of the 2016 American College of Obstetricians and Gynecologists guideline over time; and (2) to evaluate the effectiveness of aspirin for the prevention of superimposed preeclampsia and other adverse maternal and neonatal outcomes in women with chronic hypertension before and after this guideline. STUDY DESIGN This is a retrospective study of women with chronic hypertension who delivered at Thomas Jefferson University Hospital from January 2014 through June 2018. This cohort of women with chronic hypertension was divided into 2 groups, before and after the American College of Obstetricians and Gynecologists recommendation published in September 2016. Daily 81 mg of aspirin was initiated between 12 and 16 weeks. We excluded multiple gestations and incomplete records. The primary outcome was incidence of superimposed preeclampsia, and secondary outcomes were incidence of superimposed preeclampsia with or without severe features, small for gestational age, and preterm birth <37 weeks. Subgroup analysis based on risk stratification was evaluated in women with chronic hypertension requiring antihypertensive medication, history of preeclampsia, and pregestational diabetes. RESULTS We identified 457 pregnant women with chronic hypertension, 203 in the post-American College of Obstetricians and Gynecologists group and 254 in the pre-American College of Obstetricians and Gynecologists group. Aspirin 81 mg was offered to 142 (70%) in the post-American College of Obstetricians and Gynecologists group and 18 (7.0%) in the pre-American College of Obstetricians and Gynecologists group. Maternal demographics were not significantly different. The overall incidence of superimposed preeclampsia was not significantly different: 87 (34.3%) vs 72 (35.5%), P=.79, in the pre- and post-American College of Obstetricians and Gynecologists guideline groups, respectively. Superimposed preeclampsia with severe features significantly increased: 32 (12.6%) vs 9 (4.4%), P<.01, whereas superimposed preeclampsia without severe features significantly decreased: 55 (21.7%) vs 63 (31.0%), P=.03. There were no significant differences in small for gestational age neonates or preterm birth <37 weeks incidences between groups. There were no significant differences in the subgroup analysis based on the severity of chronic hypertension requiring antihypertensive medication, history of preeclampsia, or pregestational diabetes. CONCLUSION After the adoption of the American College of Obstetricians and Gynecologists guidelines in 70% of the cohort, superimposed preeclampsia, small for gestational age, and preterm birth were not significantly decreased after implementation of aspirin 81 mg initiated between 12 and 16 weeks of gestation.
Collapse
|
35
|
Fan Y, Dong Z, Zhou G, Fu J, Zhan L, Gao M, Zhu L, Zhang Y. Elevated miR-23a impairs trophoblast migration and invasiveness through HDAC2 inhibition and NF-κB activation. Life Sci 2020; 261:118358. [PMID: 32866518 DOI: 10.1016/j.lfs.2020.118358] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023]
Abstract
Preeclampsia (PE) is a pregnancy-specific disorder characterized by the onset of hypertension and proteinuria with onset after the 20th week of gestation. The pathogenesis of PE is attributed to increased trophoblast cell death and poor trophoblast migration/invasiveness. This study investigates the function of microRNA-23a (miR-23a) in PE and its effects on migration and invasion of trophoblast cells HTR-8/SVneo. We found higher expression of miR-23a in placental tissue samples from PE pregnant women compared to samples from normal pregnant women. Enhancing miR-23a expression by its specific mimic reduced HTR-8/SVneo cell migration and invasion and increased HTR-8/SVneo cell apoptosis. The dual-luciferase reporter gene assay revealed miR-23a binding with HDAC2. We found that HDAC2 was poorly expressed in placental tissue samples from PE pregnant women, and its expression correlated inversely with miR-23a expression. HTR-8/SVneo cells showed diminished HDAC2 expression upon miR-23a elevation and increased HDAC2 expression upon miR-23a inhibition. Lentivirus-mediated HDAC2 knockdown mimicked the effects of miR-23a on HTR-8/SVneo cells and led to NF-κB activation. Similarly, HDAC2 overexpression and NF-κB inhibition both abrogated the effects of miR-23a on HTR-8/SVneo cells, suggesting that miR-23a reduced HTR-8/SVneo cell migration and invasion and increased HTR-8/SVneo cell apoptosis by HDAC2 inhibition and NF-κB activation. In summary, these results support a novel role of miR-23b in invasion and apoptosis of trophoblast cells, and imply that targeting miR-23b may be a new avenue for treating PE.
Collapse
Affiliation(s)
- Yijun Fan
- Department of Obstetrics and Gynecology, The Second Hospital of Anhui Medical University, Hefei 230601, PR China.
| | - Zhen Dong
- Department of Obstetrics and Gynecology, The Second Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Guiju Zhou
- Department of Obstetrics and Gynecology, The Second Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Juanjuan Fu
- Department of Obstetrics and Gynecology, The Second Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Lei Zhan
- Department of Obstetrics and Gynecology, The Second Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Ming Gao
- Department of Obstetrics and Gynecology, The Second Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Lin Zhu
- Department of Obstetrics and Gynecology, The Second Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Yu Zhang
- Department of Obstetrics and Gynecology, The Second Hospital of Anhui Medical University, Hefei 230601, PR China.
| |
Collapse
|
36
|
Characterizing placental stiffness using ultrasound shear-wave elastography in healthy and preeclamptic pregnancies. Arch Gynecol Obstet 2020; 302:1103-1112. [PMID: 32676857 DOI: 10.1007/s00404-020-05697-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/09/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE To measure the stiffness of the placenta in healthy and preeclamptic patients in the second and third trimesters of pregnancy using ultrasound shear-wave elastography (SWE). We also aimed to evaluate the effect of age, gestational age, gravidity, parity and body mass index (BMI) on placental stiffness and a possible correlation of stiffness with perinatal outcomes. METHODS In a case-control study, we recruited a total of 47 singleton pregnancies in the second and third trimesters of which 24 were healthy and 23 were diagnosed with preeclampsia. In vivo placental stiffness was measured once at the time of recruitment for each patient. Pregnancies with posterior placentas, multiple gestation, gestational hypertension, chronic hypertension, diabetes, autoimmune disease, fetal growth restriction and congenital anomalies were excluded. RESULTS The mean placental stiffness was significantly higher in preeclamptic pregnancies compared to controls in the third trimester (difference of means = 16.8; 95% CI (9.0, 24.5); P < 0.001). There were no significant differences in placental stiffness between the two groups in the second trimester or between the severe preeclampsia and preeclampsia without severe features groups (difference of means = 9.86; 95% CI (-5.95, 25.7); P ≥ 0.05). Peripheral regions of the placenta were significantly stiffer than central regions in the preeclamptic group (difference of means = 10.67; 95% CI (0.07, 21.27); P < 0.05), which was not observed in the control group (difference of means = 0.55; 95% CI (- 5.25, 6.35); P > 0.05). We did not identify a correlation of placental stiffness with gestational age, maternal age, gravidity or parity. However, there was a statistically significant correlation with BMI (P < 0.05). In addition, pregnancies with higher placental stiffness during the 2nd and 3rd trimesters had significantly reduced birth weight (2890 ± 176 vs. 2420 ± 219 g) and earlier GA (37.8 ± 0.84 vs. 34.3 ± 0.98 weeks) at delivery (P < 0.05). CONCLUSION Compared to healthy pregnancies, placentas of preeclamptic pregnancies are stiffer and more heterogeneous. Placental stiffness is not affected by gestational age or the severity of preeclampsia but there is a correlation with higher BMI and poor perinatal outcomes.
Collapse
|
37
|
Ma XP, Liu CD, Cao GM, Zhang ZY. Transthyretin increases migration and invasion of rat placental trophoblast cells. FEBS Open Bio 2020; 10:1568-1576. [PMID: 32533762 PMCID: PMC7396443 DOI: 10.1002/2211-5463.12911] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 05/09/2020] [Accepted: 06/08/2020] [Indexed: 11/07/2022] Open
Abstract
Preeclampsia (PE) is a hypertensive disorder of pregnancy. Early diagnosis of PE is currently contingent on regular prenatal physical examinations and may be facilitated by identification of novel diagnostic markers. Transthyretin (TTR), also known as prealbumin, is primarily responsible for maintaining the normal levels of thyroxine and retinol binding protein. The expression of TTR is lower in patients with severe PE as compared with healthy controls. Here, we examined the suitability of TTR as a diagnostic marker in pregnant hypertensive rats. N'-nitro-l-arginine-methylesterhydrochloride (l-NAME) was used to generate a rat model of hypertension during pregnancy. Rat placental trophoblast cells were divided into control and TTR groups for in vitro experiments. Systolic blood pressure, diastolic blood pressure, mean blood pressure and urinary protein of hypertensive pregnant rats were higher than those of healthy pregnant rats, but these effects could be reversed by TTR treatment. There were no significant changes in blood pressure and urinary protein in healthy pregnant rats before or after TTR treatment. TTR levels in the serum and placental tissues of pregnant hypertensive rats were significantly reduced compared with those of healthy pregnant rats. Changes in placental and fetal weights in the hypertensive model could also be rescued by TTR treatment. TTR treatment significantly increased the level of matrix metalloproteinase-2/9 in hypertensive rats. Finally, in vivo and in vitro experiments demonstrated that TTR effectively increased the migration and invasion of rat placental trophoblast cells, as well as matrix metalloproteinase-2/9 levels in these cells. In conclusion, our data from a rat model suggest that TTR may have potential as a novel marker for PE diagnosis.
Collapse
Affiliation(s)
- Xiao-Peng Ma
- Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China.,Beijing Youan Hospital Affiliated to Capital Medical University, Beijing, China
| | - Chong-Dong Liu
- Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Guang-Ming Cao
- Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Zhen-Yu Zhang
- Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
38
|
Draskovic B, Nikolic T, Jacovic S, Petrovic D. Acute Kidney Damage in Pregnancy: Etiopathogenesis, Diagnostics and Basic Principles of Treatment. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2020. [DOI: 10.1515/sjecr-2017-0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Acute kidney damage associated with pregnancy occurs in 1/20.000 pregnancies. In developing countries, the main cause of the development of acute kidney damage is septic abortion, and preeclampsia in the developed countries of the world. Preeclampsia is defined as newly developed hypertension, proteinuria and swelling in pregnant women after the 20th week of gestation. It occurs due to disorders in the development of placenta and systemic disorders of the function of the endothelium of the mother. It is treated with methyldopa, magnesium sulfate and timely delivery. Urgent delivery is indicated if the age of gestation is ≥ 34 weeks. HELLP syndrome is a difficult form of preeclampsia. Its main characteristics are decreased platelet count, microangiopathic hemolysis anemia, increased concentration of aminotransferase in the serum and acute kidney damage. Severe HELLP syndrome is treated with emergency delivery, antihypertensives, magnesium sulfate, and in some cases plasmapheresis and hemodialysis. Acute fatty liver in pregnancy occurs because of decreased activity of the LCHAD enzyme of the fetus. Due to the reduced beta oxidation of fatty acids in the hepatocytes of the fetus, long chain fatty acids that cause damage to the mother’s hepatocytes are released. Swansea criteria are used for diagnosis, and the difficult form of the disease is treated with plasmapheresis and extracorporeal liver support. Atypical HUS is due to a reduced protein activity that regulates the activity of the alternative pathway of the complement system. Its main features are thrombocytopenia, microangiopathic hemolytic anemia and acute kidney damage. It is treated with plasmapheresis, and in case of resistance with eculizumab. Thrombotic thrombocytopenic purpura is due to decreased activity of the ADAMTS13 enzyme. It is characterized by thrombocytopenia, microangiopathic hemolytic anemia, high temperature, nervous system disorders and acute kidney damage. It is treated with plasmapheresis, and severe form of disease with corticosteroids and azathioprine. Early detection and timely treatment of acute kidney damage provides a good outcome for the mother and fetus.
Collapse
Affiliation(s)
- Branislava Draskovic
- Clinic of Urology, Nephrology and Dialysis, Clinical center Kragujevac , Kragujevac , Serbia
| | - Tomislav Nikolic
- Clinic of Urology, Nephrology and Dialysis, Clinical center Kragujevac , Kragujevac , Serbia
- University of Kragujevac , Faculty of Medical Sciences , Kragujevac , Serbia
| | - Sasa Jacovic
- Medicines and Medical Devices Agency of Serbia , Belgrade , Serbia
| | - Dejan Petrovic
- Clinic of Urology, Nephrology and Dialysis, Clinical center Kragujevac , Kragujevac , Serbia
- University of Kragujevac , Faculty of Medical Sciences , Kragujevac , Serbia
| |
Collapse
|
39
|
Garrido-Gomez T, Quiñonero A, Dominguez F, Rubert L, Perales A, Hajjar KA, Simon C. Preeclampsia: a defect in decidualization is associated with deficiency of Annexin A2. Am J Obstet Gynecol 2020; 222:376.e1-376.e17. [PMID: 31738896 DOI: 10.1016/j.ajog.2019.11.1250] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/31/2019] [Accepted: 11/07/2019] [Indexed: 01/09/2023]
Abstract
BACKGROUND Decidualization defects in the endometrium have been demonstrated at the time of delivery in women with severe preeclampsia and to linger for years, which suggests a maternal contribution to the pathogenesis of this condition. Global transcriptional profiling reveals alterations in gene expression, which includes down-regulation of Annexin A2 in severe preeclampsia patients with decidualization resistance. OBJECTIVE We investigated the functional role of Annexin A2 deficiency during endometrial decidualization and its potential contribution to shallow trophoblast invasion during implantation and subsequent placentation using in vitro and in vivo modeling. STUDY DESIGN Annexin A2 gene and protein levels were assessed during in vitro decidualization of human endometrial stromal cells isolated from biopsy specimens that were collected from women with previous severe preeclampsia (n=5) or normal obstetric outcomes (n=5). Next, Annexin A2 was inhibited with small interference RNA in control human endometrial stromal cells that were isolated from endometrial biopsy specimens (n=15) as an in vitro model to analyze decidualization defects at the morphologic level and the secretion of prolactin and insulin-like growth binding protein-1. Annexin A2-inhibited cells were used to evaluate motility and promotion of embryo invasion. Decidualization and placentation defects of Annexin A2 deficiency were confirmed with the use of an Annexin A2-null mouse model. RESULTS Annexin A2 gene and protein levels were down-regulated during in vitro decidualization of human endometrial stromal cells from women with previous severe preeclampsia compared with control individuals. To assess its role in the endometrial stroma, we inhibited Annexin A2 expression and detected decidualization failure as evidenced by impaired morphologic transformation, which was associated with altered actin polymerization and low prolactin and insulin-like growth binding protein-1 secretions. Functionally, in vitro models demonstrated that Annexin A2 inhibition failed to support embryo invasion. This finding was corroborated by reduced trophoblast spreading through human endometrial stromal cells, lack of motility of these cells, and reduced trophoblast invasion in the presence of conditioned media from Annexin A2-inhibited cells. Extending our discovery to an animal model, we detected that Annexin A2-null mice have a functional deficiency in decidualization and placentation that impairs fetal growth as a feature that is associated with severe preeclampsia. CONCLUSION Together, in vitro and in vivo results suggest that endometrial defects in Annexin A2 expression impair decidualization of endometrial stromal cells as well as the uterine microenvironment that promotes embryo implantation and placentation. Our findings highlight the maternal contribution to the pathogenesis of severe preeclampsia and suggest that evaluation of Annexin A2 may provide a novel strategy to assess a woman's risk of experiencing this disease and perhaps discover therapeutic interventions to improve decidualization.
Collapse
|
40
|
da Costa TX, de Almeida Pimenta Cunha MD, do Vale Bezerra PK, Azeredo FJ, Martins RR, Oliveira AG. Incidence of Adverse Drug Reactions in High-Risk Pregnancy: A Prospective Cohort Study in Obstetric Intensive Care. Eur J Clin Pharmacol 2019; 76:291-298. [PMID: 31768575 DOI: 10.1007/s00228-019-02789-9] [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: 08/05/2019] [Accepted: 10/25/2019] [Indexed: 10/25/2022]
Abstract
PURPOSE To estimate the cumulative incidence of adverse drug reactions (ADRs) in women with high-risk pregnancy hospitalized in an obstetric intensive care unit, then to describe the medicines involved and to identify major risk factors. METHODS From June 2016 to December 2017, patients admitted to the ICU with high-risk pregnancy were considered eligible in this observational, longitudinal, prospective study. Patients were investigated daily for the occurrence of ADRs through pharmaceutical anamnesis, active search in medical records and questioning of the health team. Suspected ADRs were classified according to Naranjo's algorithm. Written informed consent was obtained from all patients. Univariate and multivariate logistic regression were used to identify risk factors of ADR. RESULTS The study population consisted of 607 high-risk pregnancies from 851 women admitted to the ICU, of whom 244 admitted for non-obstetric conditions, with an ICU stay less than 24 h or readmitted to the ICU were excluded. The mean age was 27.0 ± 7.5 years-old, mean gestational age was 33.8 ± 6.3 weeks. ADR were observed in 165 women (27.2%). No severe ADR was observed and 29.7% were of moderate severity. The most often implicated medicine was magnesium sulphate (25.2%) with 44.5% of patients administered that substance experiencing ADRs consisting of somnolence (68.6%), absent patellar reflex (21.6%) and hypotension (9.8%). Risk factors of ADR were blood pressure (adjusted odds-ratio (aOR) 1.02), haemoglobin level (aOR 1.21) and body temperature (aOR 0.71). CONCLUSIONS ADRs affect about one third of high-risk pregnancies, mainly due to magnesium sulphate administrations. High blood pressure, lower body temperature, and high haemoglobin concentration on admission were associated with an increased risk of ADR.
Collapse
Affiliation(s)
- Tatiana Xavier da Costa
- Postgraduate Program in Pharmaceutical Sciences, Centro de Ciências da Saúde, Universidade Federal do Rio Grande do Norte, Natal, Brazil. .,Maternity School Januário Cicco, Centro de Ciências da Saúde, Universidade Federal do Rio Grande do Norte, Natal, Brazil. .,Faculdade de Farmácia, Centro de Ciências da Saúde, Universidade Federal do Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil.
| | - Marta Danielle de Almeida Pimenta Cunha
- Postgraduate Program in Pharmaceutical Sciences, Centro de Ciências da Saúde, Universidade Federal do Rio Grande do Norte, Natal, Brazil.,Maternity School Januário Cicco, Centro de Ciências da Saúde, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | | | | | - Rand Randall Martins
- Department of Pharmacy, Centro de Ciências da Saúde, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Antonio Gouveia Oliveira
- Department of Pharmacy, Centro de Ciências da Saúde, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
41
|
Das NS, Dheen ST, Ling EA, Bay BH, Srinivasan DK. Therapeutic Prospects in Preeclampsia - A Mini-Review. Curr Med Chem 2019; 26:4786-4798. [PMID: 30836908 DOI: 10.2174/0929867326666190228115423] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 02/07/2019] [Accepted: 02/13/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Preeclapmsia (PE) is characterized by early onset symptoms such as elevated blood pressure, proteinuria and edema in the pregnant woman, and may result in seizures in the affected female. Currently, there are no therapeutic drugs available to treat this condition, but there are interventions to regulate the symptoms based on the gestational period of the fetus, although the largely favored option is delivery of the fetus and placenta. OBJECTIVE A search for biomolecules associated with PE was conducted so as to identify diagnostic markers and therapeutic leads. RESULTS The literature search resulted in the identification of biomolecules such as Corin and Placental Protein 13 (PP13), among others that are associated with PE. Thereby, giving an insight into the various mechanistic pathways involved in the causation of PE. However, it is also evident that PE cannot be solely attributed to any single mechanism but is due to an interplay of different factors that have led to the development of this disease condition. CONCLUSION The identified biomarkers would ultimately help in understanding this complex disease and perhaps lead to the discovery of potential effective molecular targets for clinical trials, thereby providing a valuable therapeutic option for affected pregnant women.
Collapse
Affiliation(s)
- N S Das
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - S T Dheen
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - E A Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - B H Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - D K Srinivasan
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
42
|
Guo C, Cai R, Cao X, Yang Y, Wang Q, He R, An L, Peng Z, Chen Y, Ni S, Tan X, Han J, Liu X, Lin L, Yu S, Wang X, Wang C, Ma X. Deep targeted sequencing reveals the diversity of TRB-CDR3 repertoire in patients with preeclampsia. Hum Immunol 2019; 80:848-854. [PMID: 30965079 DOI: 10.1016/j.humimm.2019.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/03/2019] [Accepted: 04/06/2019] [Indexed: 12/12/2022]
Abstract
Preeclampsia (PE) is one of the major causes of maternal and perinatal mortality worldwide. This study aimed to determine the immunological characteristics of PE patients and normal pregnancy at the T cell receptor beta-chain (TRB) level by using high-throughput sequencing. High-throughput sequencing was performed to analyze the expression of TRB-CDR3 in circulating T cells. T cells were isolated from 36 healthy pregnant women, 24 patients with severe PE, and 18 patients with moderate PE. Rearranged mRNA sequences were assigned to their germline V, D, and J counterparts, and translated into proper amino acids by the IMGT database. In general, PE samples had more TRB-CDR3 reads and types than those of normal pregnant woman in the circulation, but the mean number of TRB-CDR3 reads and unique TRB-CDR3 reads in severe group was lower than that in the moderate group. In PE patients, the V7_9 and V20_1 gene loci were more prevalent than in healthy pregnant women. In addition, 4 kinds of TRB-CDR3 peptides were found to be highly relevant to the pathogenesis of PE. Of them, peptides matched to herpes simplex virus antigen-specific T cells were much lower in PE samples.
Collapse
Affiliation(s)
- Changlong Guo
- Department of Genetics, National Research Institute for Family Planning, Beijing 100081, China
| | - Ruikun Cai
- Department of Genetics, National Research Institute for Family Planning, Beijing 100081, China
| | - Xiaofang Cao
- Department of Genetics, National Research Institute for Family Planning, Beijing 100081, China
| | - Ying Yang
- Department of Genetics, National Research Institute for Family Planning, Beijing 100081, China
| | - Qidi Wang
- Department of Genetics, National Research Institute for Family Planning, Beijing 100081, China
| | - Runsheng He
- Department of Genetics, National Research Institute for Family Planning, Beijing 100081, China
| | - Lisha An
- Department of Genetics, National Research Institute for Family Planning, Beijing 100081, China
| | - Zuoqi Peng
- Department of Genetics, National Research Institute for Family Planning, Beijing 100081, China
| | - Yequn Chen
- The First Affiliated Hospital, Medical College of Shantou University, Shantou, Guangdong 515041, China
| | - Shuhua Ni
- The First Affiliated Hospital, Medical College of Shantou University, Shantou, Guangdong 515041, China
| | - Xuerui Tan
- The First Affiliated Hospital, Medical College of Shantou University, Shantou, Guangdong 515041, China
| | - Jian Han
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Xin Liu
- Department of Genetics, National Research Institute for Family Planning, Beijing 100081, China; Laboratory of Human Genetics, Beijing Hypertension League Institute, Beijing 100043, China
| | - Li Lin
- Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Song Yu
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China
| | - Xingyu Wang
- Department of Genetics, National Research Institute for Family Planning, Beijing 100081, China; Laboratory of Human Genetics, Beijing Hypertension League Institute, Beijing 100043, China.
| | - Chunlin Wang
- Stanford Genome Technology Center, Stanford University, Palo Alto CA 94003, USA.
| | - Xu Ma
- Department of Genetics, National Research Institute for Family Planning, Beijing 100081, China; Graduate School of Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
43
|
Brosens I, Brosens JJ, Muter J, Puttemans P, Benagiano G. Preeclampsia: the role of persistent endothelial cells in uteroplacental arteries. Am J Obstet Gynecol 2019; 221:219-226. [PMID: 30738027 DOI: 10.1016/j.ajog.2019.01.239] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 01/11/2019] [Accepted: 01/31/2019] [Indexed: 01/22/2023]
Abstract
We explore the potential role of the endothelial lining of uteroplacental arteries in the pathogenesis of preeclampsia, a severe pregnancy disorder characterized by incomplete invasion of the uterine vasculature by extravillous trophoblast and angiogenic imbalance. In normal pregnancy, the endothelium disappears progressively from the uteroplacental arteries and is replaced by trophoblast and deposition of fibrofibrinoid structure, underpinning the so-called physiological transformation of uterine spiral arteries. We hypothesize that partial persistence of the endothelium, albeit injured, initiates a chain of events leading to the emergence of preeclampsia in 3 sequential stages. The first stage results in retention of the endothelium in uteroplacental arteries secondary to incomplete physiological transformation of the vessels. Consequently, the uteroplacental vessels are reactive to pathological cues, which drives local arteriopathy. The second stage starts with progressive reduction in uteroplacental blood flow, generating oxidative stress in the whole placenta, and heightened maternal inflammation in response to circulating trophoblastic debris. In the third stage, generalized endotheliosis causes systemic angiogenic imbalance, hypertension, and other clinical manifestation of preeclampsia.
Collapse
|
44
|
Nielsen LH, Kronborg C, Vittinghus E, Kitlen G, Jensen BL, Knudsen UB, Ovesen PG. Is urinary excretion of plasminogen associated with development of pre-eclampsia? An observational, explorative case-control study. BMJ Open 2019; 9:e026489. [PMID: 31230006 PMCID: PMC6597096 DOI: 10.1136/bmjopen-2018-026489] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES Pre-eclampsia (PE) is characterised by renal glomerular endotheliosis and injury to the glomerular filtration barrier with proteinuria. Patients with PE display aberrant filtration of the plasma proenzyme plasminogen which is activated, in the tubular fluid, to plasmin. Plasmin may activate the epithelial sodium channel and cause impaired sodium excretion and contribute to hypertension. An explorative study was conducted to test the association between urinary total plasminogen/plasmin and the development of PE. A positive association was hypothesised. DESIGN An observational, explorative, nested case-control study of healthy pregnant women. SETTINGS A Danish County hospital. Samples were collected between 2001 and 2004. PARTICIPANTS 1631 healthy pregnant women participated. Urine samples were collected longitudinally six times during pregnancy. 30 developed PE (cases) and were compared with 146 randomly selected healthy pregnant women (controls). PRIMARY OUTCOME The association between total plasminogen/plasmin excreted in the urine and PE development is expressed by ORs. Total urinary excretion of plasminogen/plasmin was defined by the urine plasminogen-plasmin/creatinine ratio. SECONDARY OUTCOME The association between urine (u)-albumin/creatinine ratio, u-aldosterone/creatinine ratio and PE development is expressed by ORs. The correlation between urinary (u-) plasmin and u-aldosterone concentration is expressed as a correlation coefficient. RESULTS The development of PE in late pregnancy was associated with increased levels of the urine plasminogen-plasmin/creatinine ratio (OR=2.35; 95% CI: 1.12 to 4.93; p<0.05).U-aldosterone/creatinine ratio did not predict PE at any time. U-albumin/creatinine ratio was positively associated with the development of PE from gestational week 33 (OR=14.04; 95% CI: 2.56 to 76.97; p<0.01) and in week 33-35 (OR=14.15; 95% CI: 3.44 to 58.09; p<0.001) and after gestational week 36, respectively. CONCLUSION Aberrant filtration of plasminogen may contribute to the pathophysiological features of impaired sodium excretion and hypertension associated with PE late in pregnancy. However, increased urinary albumin levels reveal stronger associations with PE development compared with urinary plasminogen levels.
Collapse
Affiliation(s)
- Lise H Nielsen
- Department of Gynecology and Obstetrics, Aarhus Universitetshospital, Aarhus, Denmark
| | - Camilla Kronborg
- Department of Oncology, Aarhus Universitetshospital, Aarhus, Denmark
| | - Erik Vittinghus
- Department of Clinical Biochemistry, Regionshospitalet Viborg, Viborg, Midtjylland, Denmark
| | - Gitte Kitlen
- Department of Cardiovascular and Renal Research, Syddansk Universitet Institut for Molekylar Medicin, Odense, Denmark
| | - Boye L Jensen
- Department of Cardiovascular and Renal Research, Syddansk Universitet Institut for Molekylar Medicin, Odense, Denmark
| | - Ulla B Knudsen
- Department of Gynecology and Obstetrics, Aarhus Universitetshospital, Aarhus, Denmark
| | - Per G Ovesen
- Department of Gynecology and Obstetrics, Aarhus Universitetshospital, Aarhus, Denmark
| |
Collapse
|
45
|
Role of mammalian target of rapamycin signaling pathway in regulation of fatty acid oxidation in a preeclampsia-like mouse model treated with pravastatin. Chin Med J (Engl) 2019; 132:671-679. [PMID: 30855348 PMCID: PMC6416100 DOI: 10.1097/cm9.0000000000000129] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: Fatty acid oxidation (FAO) disorder is involved in the pathogenesis of some cases of preeclampsia (PE). Several studies show that mammalian target of rapamycin (mTOR) signaling pathway is related to FAO. Pravastatin (Pra) can promote FAO in Nω-nitro-L-arginine methyl ester (L-NAME) PE-like mouse model in our previous study. This study aimed to investigate the effect of mTOR signaling pathway in PE-like model treated with Pra. Methods: Pregnant mice were randomly injected with L-NAME as PE-like model group or saline as control group respectively, from gestational 7th to 18th day. Giving Pra (L-NAME + Pra, Control + Pra, n = 8) or normal saline (NS; L-NAME + NS, Control + NS, n = 8) from gestational 8th to 18th day, the mice were sacrificed on day 18 and their liver and placental tissues were collected. Then the activation of mTOR and its substrates in the liver and placenta were detected. And the association between mTOR activation and serum free fatty acid (FFA) levels and the expression of long-chain 3-hydroxyacyl-coenzyme A dehydrogenase (LCHAD) were evaluated using Pearson correlation test. Differences between groups were analyzed using independent t-test or one-way analysis of variance (ANOVA). Results: Both in the maternal liver and placenta, the activation of mTOR protein and its effect on substrates increased significantly in the L-NAME + NS group and decreased significantly in the L-NAME + Pra group. The p-mTOR/mTOR protein ratio decreased in the L-NAME + Pra group significantly than that in the L-NAME + NS group both in liver and placenta (liver: 0.74 ± 0.08 vs. 0.85 ± 0.06, t = 2.95, P < 0.05; placenta: 0.63 ± 0.06 vs. 0.77 ± 0.06, t = 4.64, P < 0.05). The activation of mTOR protein in the liver and placenta negatively correlated with the expression of LCHAD in the L-NAME + NS group (liver: r = −0.745, P < 0.05; placenta: r = −0.833, P < 0.05) and that in the maternal liver negatively correlated with the expression of LCHAD (r = −0.733, P < 0.05) and positively with the serum FFA levels (r = 0.841, P < 0.05) in the L-NAME + Pra group. Conclusion: The inhibition of mTOR signaling pathway might be involved in the regulation of FAO in mouse model treated with Pra.
Collapse
|
46
|
Han X, Ghaemi MS, Ando K, Peterson LS, Ganio EA, Tsai AS, Gaudilliere DK, Stelzer IA, Einhaus J, Bertrand B, Stanley N, Culos A, Tanada A, Hedou J, Tsai ES, Fallahzadeh R, Wong RJ, Judy AE, Winn VD, Druzin ML, Blumenfeld YJ, Hlatky MA, Quaintance CC, Gibbs RS, Carvalho B, Shaw GM, Stevenson DK, Angst MS, Aghaeepour N, Gaudilliere B. Differential Dynamics of the Maternal Immune System in Healthy Pregnancy and Preeclampsia. Front Immunol 2019; 10:1305. [PMID: 31263463 PMCID: PMC6584811 DOI: 10.3389/fimmu.2019.01305] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/22/2019] [Indexed: 12/11/2022] Open
Abstract
Preeclampsia is one of the most severe pregnancy complications and a leading cause of maternal death. However, early diagnosis of preeclampsia remains a clinical challenge. Alterations in the normal immune adaptations necessary for the maintenance of a healthy pregnancy are central features of preeclampsia. However, prior analyses primarily focused on the static assessment of select immune cell subsets have provided limited information for the prediction of preeclampsia. Here, we used a high-dimensional mass cytometry immunoassay to characterize the dynamic changes of over 370 immune cell features (including cell distribution and functional responses) in maternal blood during healthy and preeclamptic pregnancies. We found a set of eight cell-specific immune features that accurately identified patients well before the clinical diagnosis of preeclampsia (median area under the curve (AUC) 0.91, interquartile range [0.82-0.92]). Several features recapitulated previously known immune dysfunctions in preeclampsia, such as elevated pro-inflammatory innate immune responses early in pregnancy and impaired regulatory T (Treg) cell signaling. The analysis revealed additional novel immune responses that were strongly associated with, and preceded the onset of preeclampsia, notably abnormal STAT5ab signaling dynamics in CD4+T cell subsets (AUC 0.92, p = 8.0E-5). These results provide a global readout of the dynamics of the maternal immune system early in pregnancy and lay the groundwork for identifying clinically-relevant immune dysfunctions for the prediction and prevention of preeclampsia.
Collapse
Affiliation(s)
- Xiaoyuan Han
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Mohammad S Ghaemi
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Kazuo Ando
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Laura S Peterson
- Department of Pediatrics, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Edward A Ganio
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Amy S Tsai
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Dyani K Gaudilliere
- Department of Surgery, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Ina A Stelzer
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Jakob Einhaus
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Basile Bertrand
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Natalie Stanley
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Anthony Culos
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Athena Tanada
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Julien Hedou
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Eileen S Tsai
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Ramin Fallahzadeh
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Ronald J Wong
- Department of Pediatrics, School of Medicine, Stanford University, Palo Alto, CA, United States.,March of Dimes Prematurity Research Center, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Amy E Judy
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Maurice L Druzin
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Yair J Blumenfeld
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Mark A Hlatky
- Department of Health Research and Policy, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Cecele C Quaintance
- March of Dimes Prematurity Research Center, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Ronald S Gibbs
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Brendan Carvalho
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Gary M Shaw
- Department of Pediatrics, School of Medicine, Stanford University, Palo Alto, CA, United States.,March of Dimes Prematurity Research Center, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - David K Stevenson
- Department of Pediatrics, School of Medicine, Stanford University, Palo Alto, CA, United States.,March of Dimes Prematurity Research Center, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| |
Collapse
|
47
|
Orosz L, Orosz G, Veress L, Dosa D, Orosz L, Arany I, Fabian A, Medve L, Pap K, Karanyi Z, Toth Z, Poka R, Than NG, Torok O. Screening for preeclampsia in the first trimester of pregnancy in routine clinical practice in Hungary. J Biotechnol 2019; 300:11-19. [PMID: 31055145 DOI: 10.1016/j.jbiotec.2019.04.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/18/2019] [Accepted: 04/20/2019] [Indexed: 12/01/2022]
Abstract
We aimed to evaluate the contribution of different factors in the Fetal Medicine Foundation algorithms for preeclampsia (PE) risk calculation during first-trimester screening in Hungary. We selected subjects for the nested case-control study from a prospective cohort of 2545 low-risk pregnancies. Eighty-two patients with PE and 82 gestational age-matched controls were included. Individual PE risk was calculated using two risk-assessing softwares. Using Astraia 2.3.1, considering maternal characteristics and biophysical parameters only, detection rates (DR) were 63.6% for early-PE and 67.6% for late-PE. When we added placenta associated plasma protein A (PAPP-A) to the risk calculation, DRs decreased to 54.5% and 64.8% respectively. Using Astraia 2.8.2 with maternal characteristics and biophysical parameters resulted in the DRs of 63.6% (early-PE) and 56.3% (late-PE). If we added PAPP-A to the risk calculation, DRs improved to 72.7% and 54.9%. The addition of placental growth factor (PlGF) did not increase detection rates in either calculation. In conclusion, using maternal characteristics, biophysical parameters, and PAPP-A, an acceptable screening efficacy could be achieved for early-PE during first-trimester screening. Since PlGF did not improve efficacy in our study, we suggest setting new standard curves for PlGF in Eastern European pregnant women, and the evaluation of novel biochemical markers.
Collapse
Affiliation(s)
- Laszlo Orosz
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen Medical and Health Science Centre, Nagyerdei korut 98, 4032, Debrecen, Hungary
| | - Gergo Orosz
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen Medical and Health Science Centre, Nagyerdei korut 98, 4032, Debrecen, Hungary
| | - Lajos Veress
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen Medical and Health Science Centre, Nagyerdei korut 98, 4032, Debrecen, Hungary
| | - Diana Dosa
- Department of Family and Occupational Medicine, Faculty of Public Health and Faculty of Medicine, University of Debrecen Medical and Health Science Centre, Moricz Zsigmond krt. 22, 4032, Debrecen, Hungary
| | - Laszlo Orosz
- Departement of Obststrics and Gynaecology, Andras Josa County and Teaching Hospital, Szent Istvan ut. 68, 4400, Nyiregyhaza, Hungary.
| | - Ibolya Arany
- Departement of Neonatology, Andras Josa County and Teaching Hospital, Szent Istvan ut. 68, 4400, Nyiregyhaza, Hungary
| | - Antal Fabian
- Departement of Obststrics and Gynaecology, Andras Josa County and Teaching Hospital, Szent Istvan ut. 68, 4400, Nyiregyhaza, Hungary
| | - Laszlo Medve
- Departement of Obststrics and Gynaecology, Andras Josa County and Teaching Hospital, Szent Istvan ut. 68, 4400, Nyiregyhaza, Hungary
| | - Karoly Pap
- Departement of Obststrics and Gynaecology, Andras Josa County and Teaching Hospital, Szent Istvan ut. 68, 4400, Nyiregyhaza, Hungary
| | - Zsolt Karanyi
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen Medical and Health Science Centre, Nagyerdei korut 98, 4032, Debrecen, Hungary
| | - Zoltan Toth
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen Medical and Health Science Centre, Nagyerdei korut 98, 4032, Debrecen, Hungary
| | - Robert Poka
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen Medical and Health Science Centre, Nagyerdei korut 98, 4032, Debrecen, Hungary
| | - Nandor Gabor Than
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudosok krt. 2, 1117, Budapest, Hungary; Maternity Private Clinic of Obstetrics and Gynecology, Kiralyhago ter 8, 1126, Budapest, Hungary; First Department of Pathology and Experimental Cancer Research, Semmelweis University, Ulloi ut 26, 1085, Budapest, Hungary.
| | - Olga Torok
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen Medical and Health Science Centre, Nagyerdei korut 98, 4032, Debrecen, Hungary.
| |
Collapse
|
48
|
Affiliation(s)
- Sarosh Rana
- From the Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago, IL (S.R.)
| | - Elizabeth Lemoine
- Harvard Medical School, Boston, MA (E.L.)
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (E.L., S.A.K.)
| | - Joey P. Granger
- Department of Physiology, University of Mississippi Medical Center, Jackson (J.P.G.)
| | - S. Ananth Karumanchi
- Departments of Medicine, Obstetrics and Gynecology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (S.A.K.)
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (E.L., S.A.K.)
| |
Collapse
|
49
|
Hahn S, Hasler P, Vokalova L, van Breda SV, Lapaire O, Than NG, Hoesli I, Rossi SW. The role of neutrophil activation in determining the outcome of pregnancy and modulation by hormones and/or cytokines. Clin Exp Immunol 2019; 198:24-36. [PMID: 30768780 DOI: 10.1111/cei.13278] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2018] [Indexed: 12/16/2022] Open
Abstract
Neutrophils are often exclusively considered as a first-line innate immune defence, able to rapidly kill or trap pathogens and causing in case of over-activation tissue damage. In the female reproductive tract, however, the presence and activity of neutrophils seems to be tightly regulated. Major players in orchestrating this regulation are cyclical steroid sex hormones present during the menstrual cycle and pregnancy. This review describes the role of sex hormones in regulating directly or indirectly the functionality of neutrophils, the role of neutrophils during fertilization and pregnancy and in controlling viral, fungal and bacterial infection. This review also discusses the consequence of overt neutrophil activation in pregnancy pathologies.
Collapse
Affiliation(s)
- S Hahn
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - P Hasler
- Department of Rheumatology, Kantonsspital Aarau, Aarau, Switzerland
| | - L Vokalova
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - S V van Breda
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland.,Department of Rheumatology, Kantonsspital Aarau, Aarau, Switzerland
| | - O Lapaire
- Department of Obstetrics, University Women's Hospital Basel, Basel, Switzerland
| | - N G Than
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Center for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - I Hoesli
- Department of Obstetrics, University Women's Hospital Basel, Basel, Switzerland
| | - S W Rossi
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| |
Collapse
|
50
|
Ellis R, Katerelos M, Choy SW, Cook N, Lee M, Paizis K, Pell G, Walker S, Power DA, Mount PF. Increased expression and phosphorylation of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase isoforms in urinary exosomes in pre-eclampsia. J Transl Med 2019; 17:60. [PMID: 30819197 PMCID: PMC6394033 DOI: 10.1186/s12967-019-1806-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 02/21/2019] [Indexed: 12/13/2022] Open
Abstract
Background Glycolysis is altered in various kidney diseases, but little is known about glycolysis in pre-eclampsia, a multi-system disorder with major pathological effects on the kidney. Urinary exosomes provide a non-invasive alternative for studying changes in kidney metabolism. This study aims to characterise the expression and phosphorylation of isozymes of the key glycolytic regulatory protein, 6-phosphofructokinase-2-kinase/fructose-2,6-bisphosphatase (PFK-2/FBPase-2), in urinary exosomes of subjects with pre-eclampsia (PE), compared to normotensive non-pregnant (NC) and normotensive pregnant (NP) controls. Methods A cross-sectional study of NC (n = 19), NP (n = 23) and PE (n = 29) subjects was performed. Exosomes were isolated from urine samples by differential ultracentrifugation, and then analyzed by Western blot and densitometry for expression of PFK-2/FBPase-2 isozymes (PFKFB2, PFKFB3 and PFKFB4) and phosphorylation of PFKFB2 at residues Ser483 and Ser466 and PFKFB3 at Ser461. Results PFKFB2 expression was increased 4.7-fold in PE compared to NP (p < 0.001). PFKFB2 phosphorylation at Ser483 was increased 2.6-fold in PE compared to NP (p = 0.002). Expression of phosphorylated PFKFB2/PFKFB3 at Ser466/Ser461 was increased in PE, being present in 77.4% (95% CI 59.9–88.9%) of PE and 8.3% (95% CI 1.2–27.0%) of NP samples (p < 0.001). PFKFB3 was more commonly expressed in PE, detected in 90.3% (95% CI 74.3–97.4%) of PE and 8.3% (95% CI 1.2–27.0%) of NP samples (p < 0.001). PFKFB4 had a 7.2-fold increase in expression in PE compared to NP (p < 0.001). No significant differences between NP and NC groups were observed. Conclusion Regulatory proteins that increase glycolysis are increased in the urinary exosomes of subjects with pre-eclampsia, suggesting that renal glycolysis may be increased in this condition.
Collapse
Affiliation(s)
- R Ellis
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia.,Department of Nephrology, Austin Health, Studley Road, Heidelberg, Melbourne, VIC, 3084, Australia
| | - M Katerelos
- Department of Nephrology, Austin Health, Studley Road, Heidelberg, Melbourne, VIC, 3084, Australia.,Kidney Laboratory, Institute for Breathing and Sleep, Heidelberg, Australia
| | - S W Choy
- Department of Nephrology, Austin Health, Studley Road, Heidelberg, Melbourne, VIC, 3084, Australia
| | - N Cook
- Department of Nephrology, Austin Health, Studley Road, Heidelberg, Melbourne, VIC, 3084, Australia
| | - M Lee
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia.,Department of Nephrology, Austin Health, Studley Road, Heidelberg, Melbourne, VIC, 3084, Australia.,Kidney Laboratory, Institute for Breathing and Sleep, Heidelberg, Australia
| | - K Paizis
- Department of Nephrology, Austin Health, Studley Road, Heidelberg, Melbourne, VIC, 3084, Australia
| | - G Pell
- Mercy Hospital for Women, Heidelberg, Australia
| | - S Walker
- Mercy Hospital for Women, Heidelberg, Australia
| | - D A Power
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia.,Department of Nephrology, Austin Health, Studley Road, Heidelberg, Melbourne, VIC, 3084, Australia.,Kidney Laboratory, Institute for Breathing and Sleep, Heidelberg, Australia
| | - P F Mount
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia. .,Department of Nephrology, Austin Health, Studley Road, Heidelberg, Melbourne, VIC, 3084, Australia. .,Kidney Laboratory, Institute for Breathing and Sleep, Heidelberg, Australia.
| |
Collapse
|