1
|
Ibanoglu MC, Oskovi-Kaplan ZA, Ozgu-Erdinc AS, Kara O, Sahin D. Comparison of the Kisspeptin levels in early onset preeclampsia and late-onset preeclampsia. Arch Gynecol Obstet 2022; 306:991-996. [PMID: 35039882 DOI: 10.1007/s00404-021-06359-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 12/06/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Maternal Kisspeptin plays role in cell migration which is responsible for trophoblast invasion. We aimed to investigate the role of Kisspeptin as an invasion marker in the early-onset and late-onset preeclampsia cases. STUDY DESIGN In this case-control study, 125 patients were included: 20 patients with early-onset preeclampsia and 20 gestational-age-matched healthy controls; 45 patients with late-onset preeclampsia and 40 gestational-age-matched controls). Maternal plasma Kisspeptin concentration was measured and compared in groups regarding the presence of early-onset and late-onset preeclampsia. RESULTS In the late-onset PE group, significantly higher maternal plasma Kisspeptin values were observed compared with the control group at > 34 weeks of gestation (68.7 ± 93.4 pg/ml vs 68.5 ± 57.9 pg/ml; p = 0.004). Before the 34th week of gestation, plasma Kisspeptin levels did not show a significant difference when patients with early-onset PE and gestational-age matched controls were compared (66.8 ± 87.9 pg/ml vs 48.5 ± 91.3 pg/ml; p = 0.56). CONCLUSION Plasma Kisspeptin levels were significantly higher in women with late-onset preeclampsia, while no significant difference was observed in early-onset preeclampsia when compared with healthy gestational age-matched controls. The role of Kisspeptin proteins is still not clearly defined in the pathogenesis of preeclampsia.
Collapse
Affiliation(s)
- Mujde Can Ibanoglu
- Department of Obstetrics and Gynecology, Etlik Zubeyde Hanim Women's Health Training and Research Hospital, Ankara, Turkey
| | - Z Asli Oskovi-Kaplan
- Department of Obstetrics and Gynecology, TOBB Economy and Technical University, Ankara, Turkey.
| | - A Seval Ozgu-Erdinc
- Department of Obstetrics and Gynecology, TOBB Economy and Technical University, Ankara, Turkey
| | - Ozgur Kara
- Department of Perinatology, University of Health Sciences, Ankara City Hospital, Antakya, Hatay, Turkey
| | - Dilek Sahin
- Department of Perinatology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| |
Collapse
|
2
|
Rosenfeld CS. Placental serotonin signaling, pregnancy outcomes, and regulation of fetal brain development†. Biol Reprod 2021; 102:532-538. [PMID: 31711155 DOI: 10.1093/biolre/ioz204] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 10/09/2019] [Accepted: 10/17/2019] [Indexed: 12/31/2022] Open
Abstract
The placenta is a transient organ but essential for the survival of all mammalian species by allowing for the exchanges of gasses, nutrients, and waste between maternal and fetal placenta. In rodents and humans with a hemochorial placenta, fetal placenta cells are susceptible to pharmaceutical agents and other compounds, as they are bathed directly in maternal blood. The placenta of mice and humans produce high concentrations of serotonin (5-HT) that can induce autocrine and paracrine effects within this organ. Placental 5-HT is the primary source of this neurotransmitter for fetal brain development. Increasing number of pregnant women at risk of depression are being treated with selective serotonin-reuptake inhibitors (SSRIs) that bind to serotonin transporters (SERT), which prevents 5-HT binding and cellular internalization, allowing for accumulation of extracellular 5-HT available to bind to 5-HT(2A) receptor (5-HT(2A)R). In vitro and in vivo findings with SSRI or pharmacological blockage of the 5-HT(2A)R reveal disruptions of 5-HT signaling within the placenta can affect cell proliferation, division, and invasion. In SERT knockout mice, numerous apoptotic trophoblast cells are observed, as well as extensive pathological changes within the junctional zone. Collective data suggest a fine equilibrium in 5-HT signaling is essential for maintaining normal placental structure and function. Deficiencies in placental 5-HT may also result in neurobehavioral abnormalities. Evidence supporting 5-HT production and signaling within the placenta will be reviewed. We will consider whether placental hyposerotonemia or hyperserotonemia results in similar pathophysiological changes in the placenta and other organs. Lastly, open ended questions and future directions will be explored.
Collapse
Affiliation(s)
- Cheryl S Rosenfeld
- Bond Life Sciences Center, University of Missouri, Columbia, MO USA.,Biomedical Sciences, University of Missouri, Columbia, MO USA.,MU Informatics Institute, University of Missouri, Columbia, MO USA.,Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, MO USA.,Genetics Area Program, University of Missouri, Columbia, MO USA
| |
Collapse
|
3
|
Shaheen G, Jahan S, Bibi N, Ullah A, Faryal R, Almajwal A, Afsar T, Al-Disi D, Abulmeaty M, Al Khuraif AA, Arshad M, Razak S. Association of endothelial nitric oxide synthase gene variants with preeclampsia. Reprod Health 2021; 18:163. [PMID: 34321043 PMCID: PMC8320179 DOI: 10.1186/s12978-021-01213-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 07/21/2021] [Indexed: 12/18/2022] Open
Abstract
Background Preeclampsia (PE) is a complex pregnancy hypertensive disorder with multifaceted etiology. The endothelial nitric oxide synthase (eNOS) gene and nitric oxide (NO) levels has been reported to be associated with PE predisposition in various populations. Therefore, present study was designed to investigate the role of NO levels and eNOS gene variants in preeclamptic women in Pakistan. Methods A total of 600 women were evaluated, 188 of PE with mild features, 112 of PE with severe features and 300 normotensive pregnant women. NO levels were detected by Greiss reaction method and genotyping following sequencing was conducted for eNOS gene variants. Further insilico studies were performed to get insights into the structural and functional impact of identifies mutation on eNOS protein as well as on protein regulation. Results Reduced concentrations of NO were reported in all PE groups (p < 0.05) as compared to controls. The frequency of c.894 T (p.298Asp) and g.-786C alleles were significantly associated with PE. In addition, novel homozygous variant g.2051G > A was also significantly associated with PE when compared to normotensive women. Dynamic simulation studies revealed that Glu298Asp mutation destabilize the protein molecule and decrease the overall stability of eNOS protein. Molecular docking analysis of mutant promoter with transcription factors STAT3 and STAT6 proposed changes in protein regulation upon these reported mutations in upstream region of the gene. Conclusion Considering the results of current study, the functional alterations induced by these variants may influence the bioavailability of NO and represents a genetic risk factor for increased susceptibility to PE. However, large studies or meta-analysis are necessary to validate these findings. Supplementary Information The online version contains supplementary material available at 10.1186/s12978-021-01213-9. Preeclampsia (PE) is a complex pregnancy hypertensive disorder with multifaceted etiology characterized by increased hypertension and proteinuria after 20 weeks of gestation. The present study was directed to determine the role of eNOS in susceptibility to PE and the association of c.894G > T (p.(Glu298Asp), intron 4b/4a, g.-786 T > C and other possible variants of eNOS gene with preeclampsia in Pakistani population. Computational analysis of identified variants in the coding and non-coding region of the eNOS gene was also conducted to determine the change in gene regulation and further protein stability. A total of 600 women were evaluated, 188 with mild and 112 with PE with severe features PE with 300 normotensive pregnant women. NO levels and genotyping following sequencing was conducted for eNOS gene variants. Further insilico studies were performed to get insights into the structural and functional impact of identifies mutation on eNOS protein as well as on protein regulation. Data from the current study suggest that there might be other risk variants of the eNOS gene (g.2051G > A and g.1861G > A) and lower levels of serum NO that confers in an increased risk of PE. The detailed computational investigation further confirmed the deformities and changes in protein flexibility upon Glu298Asp. These structural alterations might be associated with preeclampsia. Variants in the promoter region of the eNOS gene further validate the change in gene regulation for the onset of disease. Identification of key structural and functional features in eNOS protein and gene regulatory region might be used for designing specific drugs for therapeutic purpose.
Collapse
Affiliation(s)
- Ghazala Shaheen
- Department of Animal Sciences, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, 45320, Pakistan
| | - Sarwat Jahan
- Department of Animal Sciences, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, 45320, Pakistan
| | - Nousheen Bibi
- Department of Bioinformatics, Shaheed Benazir Bhutto Women University, Peshawar, Pakistan
| | - Asmat Ullah
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, 45320, Pakistan
| | - Rani Faryal
- Department of Microbiology, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, 45320, Pakistan
| | - Ali Almajwal
- College of Applied Medical Sciences, Community Health Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Tayyaba Afsar
- College of Applied Medical Sciences, Community Health Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Dara Al-Disi
- College of Applied Medical Sciences, Community Health Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Mahmoud Abulmeaty
- College of Applied Medical Sciences, Community Health Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Abdulaziz Abdullah Al Khuraif
- Dental Biomaterials Research Chair, Dental Health Department, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Mohammed Arshad
- Dental Biomaterials Research Chair, Dental Health Department, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Suhail Razak
- College of Applied Medical Sciences, Community Health Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia.
| |
Collapse
|
4
|
Point-of-care ultrasound for obstetric anesthesia. Int Anesthesiol Clin 2021; 59:60-77. [PMID: 34054061 DOI: 10.1097/aia.0000000000000330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
5
|
Pi Y, Tian X, Ma J, Zhang H, Huang X. Vitamin D alleviates hypoxia/reoxygenation-induced injury of human trophoblast HTR-8 cells by activating autophagy. Placenta 2021; 111:10-18. [PMID: 34126416 DOI: 10.1016/j.placenta.2021.05.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/01/2021] [Accepted: 05/26/2021] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Attenuation of trophoblast cell dysfunction would be beneficial for retarding pre-eclampsia (PE). Vitamin D has been reported to improve trophoblast cell function in early PE, but the mechanism involved is not fully elucidated. This study is aimed to investigate whether vitamin D alleviates trophoblast cell dysfunction via regulating autophagy. METHODS Human trophoblast HTR-8 cells were cultured in hypoxia/reoxygenation (H/R) condition to simulate the oxidative stress state of early PE in vitro. MTT, Transwell and tube formation assays were respectively applied to assess cell proliferation, invasion, and angiogenesis abilities. DCFH-DA staining was performed to detect cellular reactive oxygen species levels. GFP-RFP-LC3 plasmid transfection and transmission electron microscopy were subjected to monitor autophagy. Enzyme-linked immunosorbent assay and Western blot analysis were used to detect autophagy-related and pyroptosis-associated molecules. RESULTS H/R led to severe impairments on the bio-function of HTR-8 cells, as evidenced by the deficiency of cell proliferation, invasion, and angiogenesis abilities, and the increase of cellular ROS production. Simultaneously, H/R inhibited autophagy and triggered pyroptosis. 1,25(OH)2D3, the hormonally active form of vitamin D, dramatically attenuated H/R-induced trophoblast dysfunction. Also, 1,25(OH)2D3 activated autophagy and inhibited pyroptosis. Additionally, autophagy-enhancer rapamycin exerted similar protective effect to that of 1,25(OH)2D3, whereas autophagy-inhibitor 3-methyladenine blocked the protective effect of 1,25(OH)2D3. DISCUSSION The mechanism that vitamin D alleviates trophoblast cell dysfunction is associated with autophagy induction and pyroptosis inhibition.
Collapse
Affiliation(s)
- Yalei Pi
- Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Xiaoyu Tian
- Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China.
| | - Jing Ma
- Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Huifeng Zhang
- Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Xianghua Huang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China.
| |
Collapse
|
6
|
Current Updates on Pre-eclampsia: Maternal and Foetal Cardiovascular Diseases Predilection, Science or Myth? : Future cardiovascular disease risks in mother and child following pre-eclampsia. Curr Hypertens Rep 2021; 23:16. [PMID: 33694011 DOI: 10.1007/s11906-021-01132-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Cardiovascular diseases (CVD), including pre-eclampsia (PE), remain the major cause of death and morbidity in women. This review elucidates the current knowledge, state of research and scientific information available on the post-event implications and complications of PE regarding maternal and foetal cardiovascular health. Does PE expose, predispose or aggravate a predilection to maternal and foetal CVD later in life? RECENT FINDINGS Women with a history of PE are reported to have stiffer arteries and are more likely to develop cardiovascular problems with time, especially aortic stenosis and mitral regurgitation, which were not hitherto linked with hypertensive pregnancy. Foetal cells persistence in the mother long after pregnancy, now clearly established in the lungs of mice postpartum, is suggested to portend an overexpression of STOX1, which may potentiate later life CVD. Moreover, the conventional theories of in utero stress and developmental reprogramming may not adequately explain the risk of later life CVD predilection in offspring born to mothers with pre-eclampsia as recent data has shown that siblings of offspring born from pre-eclamptic pregnancies are also at higher risk of hypertension later in life, irrespective of whether subsequent pregnancies were pre-eclamptic or normotensive. The mechanism involved in adverse cardiovascular outcome in offspring of pre-eclamptic pregnancies is most likely an intricate interaction of foetal programming, environmental and genetic factors. In light of available evidence, the question of whether PE is just a pointer or predisposing factor to maternal development of CVDs in later life begs for answers to facilitate definitive clinical solutions and preventive approaches.
Collapse
|
7
|
Zhao G, Liu J, Meng T. Oxidative stress-related genes ( EPHX1 and MnSOD) polymorphism and risk of pre-eclampsia: a meta-analysis. J Matern Fetal Neonatal Med 2021; 35:5526-5538. [PMID: 33586574 DOI: 10.1080/14767058.2021.1887123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND Previous studies have detected the association of polymorphisms in oxidative stress-related genes EPHX1 and MnSOD with pre-eclampsia (PE) risk, but the results are inconsistent among studies. Thus, a meta-analysis was performed to obtain more conclusive results. METHODS Eligible studies were retrieved in PubMed, Web of Science, EMBASE, Scopus, and CNKI. Odds ratios (ORs) with 95% confidence intervals (CIs) were utilized to evaluate the relationship between EPHX1 rs1051740, EPHX1 rs2234922, MnSOD rs4880 polymorphisms, and PE susceptibility in the genetic models. The subgroup analysis was also performed. RESULTS Fourteen studies with a total of 4250 participants were included, including 1784 PE patients and 2466 healthy women. There was a statistically significant association between EPHX1 rs1051740 polymorphism and PE in Caucasians within the allele, dominant, heterozygous, and homozygous models (OR = 0.79, 95% CI = 0.64-0.98; OR = 0.64, 95% CI = 0.47-0.87; OR = 0.61, 95% CI = 0.44-0.85; OR = 0.63, 95% CI = 0.42-0.97, respectively). There was a statistically significant association between EPHX1 rs2234922 polymorphism and PE in Middle Easterners within the recessive and homozygous models (OR = 3.59, 95% CI = 1.25-10.32; OR = 3.99, 95% CI = 1.38-11.49, respectively). There was no statistically significant association between MnSOD rs4880 polymorphism and PE within five genetic models. Subgroup analysis didn't reveal any association between MnSOD rs4880 polymorphism and PE in Asians, Caucasians, or Middle Easterners. CONCLUSIONS This meta-analysis shows a significant association between the EPHX1 rs1051740 and PE risk in Caucasians. Meantime, there was a statistically significant association between EPHX1 rs2234922 polymorphism and PE in Middle Easterners.
Collapse
Affiliation(s)
- Ge Zhao
- Department of Obstetrics, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jing Liu
- Department of Obstetrics, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Tao Meng
- Department of Obstetrics, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
8
|
Parchem JG, Kanasaki K, Lee SB, Kanasaki M, Yang JL, Xu Y, Earl KM, Keuls RA, Gattone VH, Kalluri R. STOX1 deficiency is associated with renin-mediated gestational hypertension and placental defects. JCI Insight 2021; 6:141588. [PMID: 33301424 PMCID: PMC7934881 DOI: 10.1172/jci.insight.141588] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 12/03/2020] [Indexed: 11/17/2022] Open
Abstract
The pathogenesis of preeclampsia and other hypertensive disorders of pregnancy remains poorly defined despite the substantial burden of maternal and neonatal morbidity associated with these conditions. In particular, the role of genetic variants as determinants of disease susceptibility is understudied. Storkhead-box protein 1 (STOX1) was first identified as a preeclampsia risk gene through family-based genetic linkage studies in which loss-of-function variants were proposed to underlie increased preeclampsia susceptibility. We generated a genetic Stox1 loss-of-function mouse model (Stox1 KO) to evaluate whether STOX1 regulates blood pressure in pregnancy. Pregnant Stox1-KO mice developed gestational hypertension evidenced by a significant increase in blood pressure compared with WT by E17.5. While severe renal, placental, or fetal growth abnormalities were not observed, the Stox1-KO phenotype was associated with placental vascular and extracellular matrix abnormalities. Mechanistically, we found that gestational hypertension in Stox1-KO mice resulted from activation of the uteroplacental renin-angiotensin system. This mechanism was supported by showing that treatment of pregnant Stox1-KO mice with an angiotensin II receptor blocker rescued the phenotype. Our study demonstrates the utility of genetic mouse models for uncovering links between genetic variants and effector pathways implicated in the pathogenesis of hypertensive disorders of pregnancy.
Collapse
Affiliation(s)
- Jacqueline G Parchem
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Texas, USA
| | - Keizo Kanasaki
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Soo Bong Lee
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Megumi Kanasaki
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Joyce L Yang
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Yong Xu
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Kadeshia M Earl
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rachel A Keuls
- Development, Disease Models & Therapeutics Graduate Program, Center for Cell and Gene Therapy, and Stem Cells and Regenerative Medicine Center, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Vincent H Gattone
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Li T, Wei S, Fan C, Tang D, Luo D. Nesfatin-1 Promotes Proliferation, Migration and Invasion of HTR-8/SVneo Trophoblast Cells and Inhibits Oxidative Stress via Activation of PI3K/AKT/mTOR and AKT/GSK3β Pathway. Reprod Sci 2020; 28:550-561. [PMID: 32974855 DOI: 10.1007/s43032-020-00324-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023]
Abstract
Preeclampsia (PE) is a leading cause of perinatal and maternal mortality. Considering that Nesfatin-1 was reported to be downregulated in serum of PE patients, we aimed to explore the functional role of Nesfatin-1 in trophoblast cells. Cell transfection was conducted to overexpress or inhibit Nesfatin-1, and its expression was measured by quantitative PCR. Cell proliferation, migration, and invasion abilities were determined employing CCK-8, flow cytometry, wound-healing, and transwell assays. Immunofluorescence assay was performed to detect E-cadherin and vimentin. ROS, MDA, and SOD levels were measured using their corresponding commercial kits. Western blot was used to identify the expression of vital kinases in PI3K/AKT/mTOR or GSK3β pathway and invasion-related proteins in trophoblast cells. Nesfatin-1 knockdown significantly suppressed proliferation, migration, and invasion and increased cell arrest in G1 phase, as well as downregulated expressions of MMP2/9 in HTR-8/SVneo cells. Besides, Nesfatin-1 knockdown promoted the expression of E-cadherin and reduced the expression of vimentin. Additionally, the levels of ROS, MDA, and SOD were elevated upon Nesfatin-1 knockdown. On the contrary, Nesfatin-1 overexpression exerted the opposite effects. Nesfatin-1 promoted the activation of PI3K/AKT/mTOR or GSK3β pathway, blocking of which reversed the promotive effects on trophoblast invasion and the inhibitory effects on oxidative stress of Nesfatin-1 in HTR-8/SVneo cells. In short, this study revealed that Nesfatin-1 promoted trophoblast cell proliferation, migration, invasion, and EMT and suppressed oxidative stress by activating PI3K/AKT/mTOR and AKT/GSK3β signaling pathway, laying the foundation for the development of therapeutic strategy for PE by targeting Nesfatin-1.
Collapse
Affiliation(s)
- Tingting Li
- Department of Gynaecology and Obstetrics, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, NO. 1617 Riyue Avenue, Qingyang District, Chengdu, 611731, Sichuan, China
| | - Sumei Wei
- Department of Gynaecology and Obstetrics, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, NO. 1617 Riyue Avenue, Qingyang District, Chengdu, 611731, Sichuan, China
| | - Conghong Fan
- Department of Gynaecology and Obstetrics, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, NO. 1617 Riyue Avenue, Qingyang District, Chengdu, 611731, Sichuan, China
| | - Dongmei Tang
- Department of Gynaecology and Obstetrics, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, NO. 1617 Riyue Avenue, Qingyang District, Chengdu, 611731, Sichuan, China
| | - Dan Luo
- Department of Gynaecology and Obstetrics, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, NO. 1617 Riyue Avenue, Qingyang District, Chengdu, 611731, Sichuan, China.
| |
Collapse
|
10
|
Oxidative stress and mitochondrial dysfunction in early-onset and late-onset preeclampsia. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165961. [PMID: 32916282 DOI: 10.1016/j.bbadis.2020.165961] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023]
Abstract
Preeclampsia is a pregnancy-specific syndrome with multisystem involvement which leads to foetal, neonatal, and maternal morbidity and mortality. This syndrome is characterized by the onset of clinical signs and symptoms and delivery before (early-onset preeclampsia, eoPE), or after (late-onset preeclampsia, loPE), the 34 weeks of gestation. Preeclampsia is a mitochondrial disorder where its differential involvement in eoPE and loPE is unclear. Mitochondria regulate cell metabolism and are a significant source of reactive oxygen species (ROS). The syncytiotrophoblast in eoPE and loPE show altered mitochondrial structure and function resulting in ROS overproduction, oxidative stress, and cell damage and death. Mitochondrial dysfunction in eoPE may result from altered expression of several molecules, including dynamin-related protein 1 and mitofusins, compared with loPE where these factors are either reduced or unaltered. Equally, mitochondrial fusion/fission dynamics seem differentially modulated in eoPE and loPE. It is unclear whether the electron transport chain and oxidative phosphorylation are differentially altered in these two subgroups of preeclampsia. However, the activity of complex IV (cytochrome c oxidase) and the expression of essential proteins involved in the electron transport chain are reduced, leading to lower oxidative phosphorylation and mitochondrial respiration in the preeclamptic placenta. Interventional studies in patients with preeclampsia using the coenzyme Q10, a key molecule in the electron transport chain, suggest that agents that increase the antioxidative capacity of the placenta may be protective against preeclampsia development. In this review, the mitochondrial dysfunction in both eoPE and loPE is summarized. Therapeutic approaches are discussed in the context of contributing to the understanding of mitochondrial dysfunction in eoPE and loPE.
Collapse
|
11
|
Tong J, Niu Y, Chen ZJ, Zhang C. Comparison of the transcriptional profile in the decidua of early-onset and late-onset pre-eclampsia. J Obstet Gynaecol Res 2020; 46:1055-1066. [PMID: 32281216 DOI: 10.1111/jog.14257] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/28/2020] [Accepted: 03/16/2020] [Indexed: 12/14/2022]
Abstract
AIM To compare early-onset pre-eclampsia (EOPE) and late-onset pre-eclampsia (LOPE) and provide insight into the pathophysiology of pre-eclampsia (PE). METHODS Our recent work compared the transcriptomics in decidua of EOPE, LOPE and normal pregnancies (NP). RESULTS We found there are a significant number of genes uniquely expressed in the decidua of EOPE and LOPE comparing with NP. Moreover, EOPE and LOPE have their distinct profiles. Unique EOPE-associated genes were mainly involved in apoptosis related pathways such as 'apoptosis' and 'Ras signaling pathway'. PIK3CB and BCL-2 are the core regulatory genes in EOPE decidua, their abnormal expression caused decidual abnormal apoptosis which is relevant to the pathogenesis of EOPE. Whereas, LOPE is a more complicated entity which has more special LOPE-associated genes involved in decidua differentiation, especially in 'gap junction pathway', 'vascular smooth muscle contraction' and 'long-term depression'. PIK3CB, FLT1, CBLC and ITGA7 are the core regulatory genes differentially expressed in EOPE decidua comparing with LOPE. CONCLUSION In brief, the different decidual transcriptomics of EOPE and LOPE may correlate with their different etiology. These findings highlight the complex pathophysiology of PE and provide potential targets for a new treatment strategy in patients with PE.
Collapse
Affiliation(s)
- Jing Tong
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yichao Niu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Cong Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 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, Ji'nan, China
| |
Collapse
|
12
|
Mtshali Z, Moodley J, Naicker T. An Insight into the Angiogenic and Lymphatic Interplay in Pre-eclampsia Comorbid with HIV Infection. Curr Hypertens Rep 2020; 22:35. [PMID: 32200445 DOI: 10.1007/s11906-020-01040-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW To provide insight on the imbalance of angiogenic and lymphangiogenic factors in pre-eclampsia, as well as highlight polymorphism in genes related to angiogenesis and lymphangiogenesis. RECENT FINDINGS The pregnancy-specific disorder pre-eclampsia is diagnosed by the presence of hypertension with/without proteinuria, after 20 weeks of gestation. The pathogenesis of pre-eclampsia remains ambiguous, but research over the years has identified an imbalance in maternal and foetal factors. Familial predisposition and gene variation are also linked to pre-eclampsia development. The sFlt-1/PIGF ratio has attracted great attention over the years; more recently several researchers have reported that a sFlt-1/PIGF ratio of ≤ 38 can be used to predict short-term absence of pre-eclampsia. This ratio has the potential to prevent adverse pregnancy outcomes and reduce healthcare costs significantly. Genome-wide studies have additionally identified variation in the foetal gene near Flt-1. The development of preeclampsia is not limited to the maternal interface, but foetal involvement as well as genetic interplay is associated with the disorder.
Collapse
Affiliation(s)
- Zamahlabangane Mtshali
- Optics and Imaging Centre, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa.
| | - Jagidesa Moodley
- Department of Obstetrics and Gynaecology and Women's Health and HIV Research Group, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
13
|
Rosenfeld CS. The placenta-brain-axis. J Neurosci Res 2020; 99:271-283. [PMID: 32108381 DOI: 10.1002/jnr.24603] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/25/2020] [Accepted: 02/12/2020] [Indexed: 12/18/2022]
Abstract
All mammalian species depend on the placenta, a transient organ, for exchange of gases, nutrients, and waste between the mother and conceptus. Besides serving as a conduit for such exchanges, the placenta produces hormones and other factors that influence maternal physiology and fetal development. To meet all of these adaptations, the placenta has evolved to become the most structurally diverse organ within all mammalian taxa. However, commonalities exist as to how placental responses promote survival against in utero threats and can alter the trajectory of fetal development, in particular the brain. Increasing evidence suggests that reactions of the placenta to various in utero stressors may lead to long-standing health outcomes, otherwise considered developmental origin of health and disease effects. Besides transferring nutrients and gases, the placenta produces neurotransmitters, including serotonin, dopamine, norepinephrine/epinephrine, that may circulate and influence brain development. Neurobehavioral disorders, such as autism spectrum disorders, likely trace their origins back to placental disturbances. This intimate relationship between the placenta and brain has led to coinage of the term, the placenta-brain-axis. This axis will be the focus herein, including how conceptus sex might influence it, and technologies employed to parse out the effects of placental-specific transcript expression changes on later neurobehavioral disorders. Ultimately, the placenta might provide a historical record of in utero threats the fetus confronted and a roadmap to understand how placenta responses to such encounters impacts the placental-brain-axis. Improved early diagnostic and preventative approaches may thereby be designed to mitigate such placental disruptions.
Collapse
Affiliation(s)
- Cheryl S Rosenfeld
- Biomedical Sciences, University of Missouri, Columbia, MO, USA.,Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.,MU Informatics Institute, University of Missouri, Columbia, MO, USA.,Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, MO, USA.,Genetics Area Program, University of Missouri, Columbia, MO, USA
| |
Collapse
|
14
|
Ding L, Blitz MJ, Wing DA, Epstein AJ, Gjessing HK, Wilson ML. PHLDA2 gene polymorphisms and risk of HELLP syndrome and severe preeclampsia. Pregnancy Hypertens 2020; 19:190-194. [PMID: 32062476 DOI: 10.1016/j.preghy.2020.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 01/21/2020] [Accepted: 01/26/2020] [Indexed: 10/25/2022]
Abstract
OBJECTIVE Pleckstrin homology-like domain, family A, member 2 (PHLDA2) is a maternally expressed imprinted gene. Loss of imprinting in PHLDA2 is associated with abnormal placental development and fetal growth restriction. Our objective was to determine whether genetic variation in PHLDA2 is also associated with risk of HELLP syndrome and preeclampsia (PE) with severe features. STUDY DESIGN Case (n = 162) and control (n = 33) mother-father-child triads were recruited using an internet-based method. Medical records were reviewed to verify clinical diagnosis of self-reported cases. DNA was genotyped for three polymorphisms in the PHLDA2 gene using TaqMan assays: rs13390, rs1056819, rs2583435. MAIN OUTCOME MEASURES To examine the association between minor alleles and haplotypes with HELLP syndrome and PE with severe features, relative risks and 95% confidence intervals were estimated using log-linear models, adjusting for the correlation between familial genotypes, using HAPLIN. RESULTS There was no association identified between PHLDA2 gene polymorphisms or haplotypes and HELLP syndrome and PE with severe features. No parent-of-origin effects were observed. CONCLUSION Genetic variation in the PHLDA2 gene is not associated with HELLP syndrome or PE with severe features.
Collapse
Affiliation(s)
- Li Ding
- Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Matthew J Blitz
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, North Shore University Hospital, Manhasset, NY, USA
| | - Deborah A Wing
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of California, Irvine, Orange, CA, USA
| | - Aaron J Epstein
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Håkon K Gjessing
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway; Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Melissa L Wilson
- Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Benny P, Yamasato K, Yunits B, Zhu X, Ching T, Garmire LX, Berry MJ, Towner D. Maternal cardiovascular-related single nucleotide polymorphisms, genes, and pathways associated with early-onset preeclampsia. PLoS One 2019; 14:e0222672. [PMID: 31557190 PMCID: PMC6762142 DOI: 10.1371/journal.pone.0222672] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 09/04/2019] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Preeclampsia is a medical condition complicated with hypertension and proteinuria during pregnancy. While preeclampsia affects approximately 5% of pregnancies, it remains without a cure. In addition, women who had preeclampsia during pregnancy have been reported to have an increased risk for cardiovascular disease later in life. However, the disease etiology and molecular mechanisms remain poorly understood. The paucity in the literature on preeclampsia associated maternal cardiovascular risk in different ethnic populations also present a need for more research. Therefore, the objective of this study was to identify cardiovascular/metabolic single nucleotide polymorphisms (SNPs), genes, and regulatory pathways associated with early-onset preeclampsia. MATERIALS AND METHODS We compared maternal DNAs from 31 women with early-onset preeclampsia with those from a control group of 29 women without preeclampsia who delivered full-term normal birthweight infants. Women with multiple gestations and/or known medical disorders associated with preeclampsia (pregestational diabetes, chronic hypertension, renal disease, hyperthyroidism, and lupus) were excluded. The MetaboChip genotyping array with approximately 197,000 SNPs associated with metabolic and cardiovascular traits was used. Single nucleotide polymorphism analysis was performed using the SNPAssoc program in R. The Truncated Product Method was used to identify significantly associated genes. Ingenuity Pathway Analysis and Ingenuity Causal Network Analysis were used to identify significantly associated disease processes and regulatory gene networks respectively. RESULTS The early-onset preeclampsia group included 45% Filipino, 26% White, 16% other Asian, and 13% Native Hawaiian and other Pacific Islanders, which did not differ from the control group. There were no SNPs associated with early-onset preeclampsia after correction for multiple comparisons. However, through gene-based tests, 68 genes and 23 cardiovascular disease-related processes were found to be significantly associated. Associated gene regulatory networks involved cellular movement, cardiovascular disease, and inflammatory disease. CONCLUSIONS Multiple cardiovascular genes and diseases demonstrate associations with early-onset preeclampsia. This unfolds new areas of research regarding the genetic determinants of early-onset preeclampsia and their relation to future cardiovascular disease.
Collapse
Affiliation(s)
- Paula Benny
- University of Hawaii Cancer Center, Honolulu, Hawai’i, United States of America
| | - Kelly Yamasato
- Department of Obstetrics, Gynecology, and Women’s Health, John A Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawai’i, United States of America
| | - Breck Yunits
- University of Hawaii Cancer Center, Honolulu, Hawai’i, United States of America
| | - Xun Zhu
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, Hawai’i, United States of America
| | - Travers Ching
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, Hawai’i, United States of America
| | - Lana X. Garmire
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Marla J. Berry
- Department of Cell and Molecular Biology, John A Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawai’i, United States of America
| | - Dena Towner
- Department of Obstetrics, Gynecology, and Women’s Health, John A Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawai’i, United States of America
| |
Collapse
|
16
|
Örgül G, Aydın Haklı D, Özten G, Fadiloğlu E, Tanacan A, Beksaç MS. First trimester complete blood cell indices in early and late onset preeclampsia. Turk J Obstet Gynecol 2019; 16:112-117. [PMID: 31360585 PMCID: PMC6637787 DOI: 10.4274/tjod.galenos.2019.93708] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/20/2019] [Indexed: 12/01/2022] Open
Abstract
Objective This study aimed to compare the first trimester complete blood count (CBC) indices of pregnancies complicated by early-onset preeclampsia (EOPE) or late-onset preeclampsia (LOPE). Material and Methods A retrospective case-control study was conducted with 186 patients. Patients were classified into three subgroups: EOPE, LOPE, and control groups. First trimester CBC results were obtained for each patient. Hemoglobin, hematocrit, red blood cell distribution width, mean corpuscular volume, white blood cell (WBC) count, neutrophil, eosinophil, basophil, lymphocyte, monocyte, mean platelet volume, platelet distribution width, plateletcrit, and platelet count were compared. The neutrophil lymphocyte ratio was calculated by dividing the absolute lymphocyte count by the absolute neutrophil count. The platelet lymphocyte ratio was calculated by dividing the absolute lymphocyte count by the absolute platelet count. Results The total number of cases was 21, 42, and 123, in the EOPE, LOPE, and control groups, respectively. There were statistically significant differences in the total WBC and neutrophil counts between the three groups (both p<0.05). WBC and neutrophil counts were found to be highest in the EOPE group, and the LOPE group had higher levels compared with controls. The optimal cut-off values to predict EOPE for WBC and neutrophil counts were 9.55×103/ μL (sensitivity 71.4% and specificity 70.7%) and 6.45×103/μL (sensitivity 66.7% and specificity 74.8%), respectively. Conclusion Increased first trimester WBC and neutrophil counts may be predictive for EOPE.
Collapse
Affiliation(s)
- Gökçen Örgül
- Hacettepe University Faculty of Medicine, Department of Obstetrics and Gynecology, Division of Perinatology, Ankara, Turkey
| | - Duygu Aydın Haklı
- Hacettepe University Faculty of Medicine, Department of Biostatistics, Ankara, Turkey
| | - Gonca Özten
- Hacettepe University Faculty of Medicine, Department of Obstetrics and Gynecology, Division of Perinatology, Ankara, Turkey
| | - Erdem Fadiloğlu
- Hacettepe University Faculty of Medicine, Department of Obstetrics and Gynecology, Division of Perinatology, Ankara, Turkey
| | - Atakan Tanacan
- Hacettepe University Faculty of Medicine, Department of Obstetrics and Gynecology, Division of Perinatology, Ankara, Turkey
| | - Mehmet Sinan Beksaç
- Hacettepe University Faculty of Medicine, Department of Obstetrics and Gynecology, Division of Perinatology, Ankara, Turkey
| |
Collapse
|
17
|
Chen J, Yue C, Xu J, Zhan Y, Zhao H, Li Y, Ye Y. Downregulation of receptor tyrosine kinase-like orphan receptor 1 in preeclampsia placenta inhibits human trophoblast cell proliferation, migration, and invasion by PI3K/AKT/mTOR pathway accommodation. Placenta 2019; 82:17-24. [PMID: 31174622 DOI: 10.1016/j.placenta.2019.05.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 05/02/2019] [Accepted: 05/07/2019] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Invasive deficiency of the trophoblast and poor remodeling of the uterine spiral arteries were probably the primary pathogenesis causes of preeclampsia (PE). The expression of receptor tyrosine kinase-like orphan receptor 1 (ROR1) during embryogenesis had been previously confirmed and was closely related to the function of tumor cells, which was similar to the characteristics of trophoblasts. In this work, we investigated the expression profile of ROR1 in preeclampsia placentas and the functional role of ROR1 in trophoblast cells, as well as the associated molecular mechanisms. METHODS The localization expression of ROR1 in the placenta was detected by immunohistochemistry in 20 cases of normal term pregnancy, preterm delivery, late-onset severe PE, and early-onset severe PE, respectively. The expression levels were determined by fluorescence quantitative PCR and Western blot. The influence of ROR1 on trophoblast proliferation, migration, invasion, and potential regulatory pathways was evaluated in HTR-8/SVneo cell lines by transient transfection methods. RESULTS The levels of ROR1 in the placental tissues in PE were significantly lower than those in normal term pregnancy and preterm delivery. Moreover, the expression levels of ROR1 in early-onset severe PE were significantly lower than those in its late counterparts. ROR1 overexpression increased cell proliferation, migration, and invasion of HTR-8/SVneo cells, whereas its silencing had the opposite effect. Meanwhile, the phosphorylation levels of critical kinases in the PI3K/AKT/mTOR pathways were increased by ROR1 overexpression, whereas they were decreased by the silencing of ROR1. CONCLUSION ROR1 might be involved in the development of PE through regulating trophoblast viability, migration, and invasion by PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Jie Chen
- Department of Obstetrics and Gynecology, Qingdao University, Qingdao, 266000, China; Department of Obstetrics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Chongyu Yue
- Department of Obstetrics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Jine Xu
- Department of Obstetrics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Ying Zhan
- Department of Obstetrics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Han Zhao
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yan Li
- Department of Obstetrics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yuanhua Ye
- Department of Obstetrics and Gynecology, Qingdao University, Qingdao, 266000, China; Department of Obstetrics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
18
|
Yang X, Bian Y, Wan J, Li L, Yang P, Zhao S, Zhao H. Variants in the 5'‐UTR of
APELA
gene in women with preeclampsia. Prenat Diagn 2019; 39:308-313. [PMID: 30719741 DOI: 10.1002/pd.5431] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 12/13/2018] [Accepted: 01/30/2019] [Indexed: 01/12/2023]
Affiliation(s)
- Xin Yang
- Center for Reproductive MedicineShandong Provincial Hospital Affiliated to Shandong University Jinan China
- Reproductive Medicine Center of Zibo Maternity and Child Health Hospital Zibo China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University Jinan China
- The Key Laboratory for Reproductive EndocrinologyShandong University, Ministry of Education Jinan China
| | - Yuehong Bian
- Center for Reproductive MedicineShandong Provincial Hospital Affiliated to Shandong University Jinan China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University Jinan China
- The Key Laboratory for Reproductive EndocrinologyShandong University, Ministry of Education Jinan China
| | - Jipeng Wan
- Department of Obstetrics and GynecologyShandong Provincial Hospital Affiliated to Shandong University Jinan China
| | - Lei Li
- Center for Reproductive MedicineShandong Provincial Hospital Affiliated to Shandong University Jinan China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University Jinan China
- The Key Laboratory for Reproductive EndocrinologyShandong University, Ministry of Education Jinan China
- Department of Biological SciencesUniversity of Notre Dame Notre Dame IN USA
| | - Ping Yang
- Center for Reproductive MedicineShandong Provincial Hospital Affiliated to Shandong University Jinan China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University Jinan China
- The Key Laboratory for Reproductive EndocrinologyShandong University, Ministry of Education Jinan China
| | - Shigang Zhao
- Center for Reproductive MedicineShandong Provincial Hospital Affiliated to Shandong University Jinan China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University Jinan China
- The Key Laboratory for Reproductive EndocrinologyShandong University, Ministry of Education Jinan China
| | - Han Zhao
- Center for Reproductive MedicineShandong Provincial Hospital Affiliated to Shandong University Jinan China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University Jinan China
- The Key Laboratory for Reproductive EndocrinologyShandong University, Ministry of Education Jinan China
| |
Collapse
|
19
|
Abstract
Preeclampsia, a leading cause of maternal and perinatal morbidity and mortality worldwide, is accompanied by shallow placentation and deficient remodeling of the uterine spiral arteries by invasive placental trophoblast cells during the first trimester of pregnancy. Here, we generated induced pluripotent stem cells from umbilical cords of normal pregnancies and ones complicated by early onset preeclampsia (EOPE) and converted them to trophoblast to recapitulate events of early pregnancy. Parameters disturbed in EOPE, including trophoblast invasiveness, were assessed. Under low O2, both sets of cells behaved similarly, but, under the more stressful 20% O2 conditions, the invasiveness of EOPE trophoblast was markedly reduced. Gene expression changes in EOPE trophoblast suggested a dysregulation invasion linked to high O2. We describe a model for early onset preeclampsia (EOPE) that uses induced pluripotent stem cells (iPSCs) generated from umbilical cords of EOPE and control (CTL) pregnancies. These iPSCs were then converted to placental trophoblast (TB) representative of early pregnancy. Marker gene analysis indicated that both sets of cells differentiated at comparable rates. The cells were tested for parameters disturbed in EOPE, including invasive potential. Under 5% O2, CTL TB and EOPE TB lines did not differ, but, under hyperoxia (20% O2), invasiveness of EOPE TB was reduced. RNA sequencing analysis disclosed no consistent differences in expression of individual genes between EOPE TB and CTL TB under 20% O2, but, a weighted correlation network analysis revealed two gene modules (CTL4 and CTL9) that, in CTL TB, were significantly linked to extent of TB invasion. CTL9, which was positively correlated with 20% O2 (P = 0.02) and negatively correlated with invasion (P = 0.03), was enriched for gene ontology terms relating to cell adhesion and migration, angiogenesis, preeclampsia, and stress. Two EOPE TB modules, EOPE1 and EOPE2, also correlated positively and negatively, respectively, with 20% O2 conditions, but only weakly with invasion; they largely contained the same sets of genes present in modules CTL4 and CTL9. Our experiments suggest that, in EOPE, the initial step precipitating disease is a reduced capacity of placental TB to invade caused by a dysregulation of O2 response mechanisms and that EOPE is a syndrome, in which unbalanced expression of various combinations of genes affecting TB invasion provoke disease onset.
Collapse
|
20
|
Huang L, Li Y, Wang C, Li N, Hou Y, Chang R, Sun M, Wang R, Zhu L, Qiao C. Overexpression of Collapsin Response Mediator Protein 1 Inhibits Human Trophoblast Cells Proliferation, Migration, and Invasion. Reprod Sci 2018; 26:954-960. [PMID: 30466368 DOI: 10.1177/1933719118799214] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Collapsin response mediator protein 1 (CRMP-1) is widely expressed in the nervous system and has tumor suppressive effects. Our previous studies have demonstrated that CRMP-1 was expressed in the trophoblasts of the whole stage of pregnancy with significantly increasing expression in the placenta of early-onset preeclampsia. Preeclampsia, especially early onset, is strongly associated with the dysfunction of trophoblast including proliferation, apoptosis, migration, and invasion. In this study, we found an inhibitory effect of CRMP-1 on proliferation, migration, invasion, and an enhanced effect on apoptosis in human trophoblast cell lines HTR-8/SVneo and JEG-3 by MTT assay, colony formation assay, cell viability assay, caspase 3/7 activity assay, scratch wound assay, and Matrigel Transwell assay. Overexpression of CRMP-1 in trophoblast cells led to downregulate expression of matrix metalloproteinase 2 and 9. The expression of CRMP-1 was detected by real-time quantitative polymerase chain reaction and Western blot analysis. Thus, we suggested that CRMP-1 might have implications for the pathogenesis of preeclampsia by regulating the biological behavior of trophoblast cells.
Collapse
Affiliation(s)
- Ling Huang
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, No.36, Sanhao street, Shenyang, Liaoning Province, 110004, China.,Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| | - Yuanyuan Li
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, No.36, Sanhao street, Shenyang, Liaoning Province, 110004, China.,Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| | - Chunhui Wang
- Department of Hepatobiliary Surgery, General Hospital of Shenyang Military Region, Shenyang, Liaoning Province, China
| | - Na Li
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, No.36, Sanhao street, Shenyang, Liaoning Province, 110004, China.,Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China.,Department of Obstetrics and Gynecology, Shenyang Forth People's Hospital, Shenyang, Liaoning Province, China
| | - Yue Hou
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, No.36, Sanhao street, Shenyang, Liaoning Province, 110004, China.,Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| | - Ruijing Chang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Man Sun
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, No.36, Sanhao street, Shenyang, Liaoning Province, 110004, China.,Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| | - Ruochen Wang
- Department of Biomedical Engineering, University of Wisconsin at Madison, Madison, WI, USA
| | - Lei Zhu
- Shenyang Academy of Environmental Sciences, Shenyang, Liaoning Province, China
| | - Chong Qiao
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, No.36, Sanhao street, Shenyang, Liaoning Province, 110004, China. .,Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China. .,Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
21
|
Pregnancy Outcome in Women with Obstetric and Thrombotic Antiphospholipid Syndrome-A Retrospective Analysis and a Review of Additional Treatment in Pregnancy. Clin Rev Allergy Immunol 2018; 53:54-67. [PMID: 27395067 DOI: 10.1007/s12016-016-8569-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Antiphospholipid syndrome (APS) is associated with pregnancy complications such as recurrent early fetal loss (RFL), fetal death, preeclampsia (PE), and intrauterine growth restriction (obstetric APS/OAPS). Other clinical manifestations are venous and/or arterial thromboses (thrombotic APS/TAPS). The data of 37 pregnancies with OAPS and 37 pregnancies with TAPS were analyzed and compared. Overall, the most frequent APS antibodies (aPl) were LA as well as "triple-positivity"; LA antibodies were significantly more frequent in women with TAPS (67.6 % TAPS vs. 29.7 % OAPS, p < 0.010), whereas "triple-positivity" was significantly more seen in women with OAPS (40.5 % OAPS vs. 13.5 % TAPS, p < 0.010). Adequate therapy has been administered in nearly all pregnancies with TAPS, whereas in 18.9 % of pregnancies with OPS, no therapy has been given at all. One woman in OAPS and four women in TAPS were treated with plasmapheresis and immunoadsorption. There was no significant association between adverse obstetric outcome and therapy. The most frequent pregnancy complications were RFL in the OAPS group (32.4 vs. 13.5 % in TAPS) and PE in the TAPS group (18.9 % in OAPS and TAPS, respectively). The data of our study showed that pregnancies with OAPS and TAPS have a similar rate of pregnancy complications. However, pregnancies with OAPS tend to have rather RFL. Although we were not able to reveal a significant association with adverse obstetric outcome, it seems that the current adequate therapy for APS in pregnancy, consisting of LDA and LMWH, might rather prevent the development of RFL. Additionally, it might be considered to divide the obstetric APS into obstetric APS with early pregnancy complications and obstetric APS with late pregnancy complications. The division into two groups of obstetric APS might facilitate the choice of additional therapy in these women.
Collapse
|
22
|
Endler M, Cnattingius S, Granfors M, Wikström AK. The inherited risk of retained placenta: a population based cohort study. BJOG 2017; 125:737-744. [PMID: 28731581 DOI: 10.1111/1471-0528.14828] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2017] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To investigate whether retained placenta in the first generation is associated with an increased risk of retained placenta in the second generation. DESIGN Population-based cohort study. SETTING Sweden. POPULATION Using linked generational data from the Swedish Medical Birth Register 1973-2012, we identified 494 000 second-generation births with information on the birth of the mother (first-generation index birth). For 292 897 of these births there was information also on the birth of the father. METHODS Risk of retained placenta in the second generation was calculated as adjusted odds ratios (aOR) by unconditional logistic regression with 95% confidence intervals (95% CI) according to whether retained placenta occurred in a first generation birth or not. MAIN OUTCOME Retained placenta in the second generation. RESULTS The risk of retained placenta in a second-generation birth was increased if retained placenta had occurred at the mother's own birth (aOR 1.66, 95% CI 1.52-1.82), at the birth of one of her siblings (aOR 1.58, 95% CI 1.43-1.76) or both (aOR 2.75, 95% CI 2.18-3.46). The risk was slightly increased if retained placenta had occurred at the birth of the father (aOR 1.23, 95% CI 1.07-1.41). For preterm births in both generations, the risk of retained placenta in the second generation was increased six-fold if retained placenta had occurred at the mother's birth (OR 6.55, 95% CI 2.68-16.02). CONCLUSION There is an intergenerational recurrence of retained placenta on the maternal and most likely also on the paternal side. The recurrence risk seems strongest in preterm pregnancies. TWEETABLE ABSTRACT A population-based cohort study suggests that there is an intergenerational recurrence of retained placenta.
Collapse
Affiliation(s)
- M Endler
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, Sweden
| | - S Cnattingius
- Department of Medicine, Clinical Epidemiology Unit, Karolinska Institutet, Stockholm, Sweden
| | - M Granfors
- Department of Medicine, Clinical Epidemiology Unit, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Sciences, Karolinska Institutet, Danderyd, Sweden
| | - A-K Wikström
- Department of Medicine, Clinical Epidemiology Unit, Karolinska Institutet, Stockholm, Sweden.,Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| |
Collapse
|
23
|
Perales A, Delgado JL, de la Calle M, García‐Hernández JA, Escudero AI, Campillos JM, Sarabia MD, Laíz B, Duque M, Navarro M, Calmarza P, Hund M, Álvarez FV. sFlt-1/PlGF for prediction of early-onset pre-eclampsia: STEPS (Study of Early Pre-eclampsia in Spain). ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2017; 50:373-382. [PMID: 27883242 PMCID: PMC5836987 DOI: 10.1002/uog.17373] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 11/08/2016] [Accepted: 11/11/2016] [Indexed: 05/31/2023]
Abstract
OBJECTIVE A high ratio of soluble fms-like tyrosine kinase-1 (sFlt-1) to placental growth factor (PlGF) has been linked to pre-eclampsia (PE). We evaluated the sFlt-1/PlGF ratio as a predictive marker for early-onset PE in women at risk of PE. METHODS This prospective, Spanish, multicenter study included pregnant women with a risk factor for PE, including intrauterine growth restriction, PE, eclampsia or hemolysis, elevated liver enzymes and low platelet count syndrome in previous pregnancy, pregestational diabetes or abnormal uterine artery Doppler. The primary objective was to show that the sFlt-1/PlGF ratio at 20, 24 and 28 weeks' gestation was predictive of early-onset PE (< 34 + 0 weeks). Serum sFlt-1 and PlGF were measured at 20, 24 and 28 weeks. Multivariate logistic regression was used to develop a predictive model. RESULTS A total of 819 women were enrolled, of which 729 were suitable for analysis. Of these, 78 (10.7%) women developed PE (24 early onset and 54 late onset). Median sFlt-1/PlGF ratio at 20, 24 and 28 weeks was 6.3 (interquartile range (IQR), 4.1-9.3), 4.0 (IQR, 2.6-6.3) and 3.3 (IQR, 2.0-5.9), respectively, for women who did not develop PE (controls); 14.5 (IQR, 5.5-43.7), 18.4 (IQR, 8.2-57.9) and 51.9 (IQR, 11.5-145.6) for women with early-onset PE; and 6.7 (IQR, 4.6-9.9), 4.7 (IQR, 2.8-7.2) and 6.0 (IQR, 3.8-10.5) for women with late-onset PE. Compared with early-onset PE, the sFlt-1/PlGF ratio was significantly lower in controls (P < 0.001 at each timepoint) and in women with chronic hypertension (P < 0.001 at each timepoint), gestational hypertension (P < 0.001 at each timepoint) and late-onset PE (P < 0.001 at each timepoint). A prediction model for early-onset PE was developed, which included the sFlt-1/PlGF ratio plus mean arterial pressure, being parous and previous PE, with areas under the receiver-operating characteristics curves of 0.86 (95% CI, 0.77-0.95), 0.91 (95% CI, 0.85-0.97) and 0.93 (95% CI, 0.86-0.99) at 20, 24 and 28 weeks, respectively, and was superior to models using the sFlt-1/PlGF ratio alone or uterine artery mean pulsatility index. CONCLUSIONS The sFlt-1/PlGF ratio can improve prediction of early-onset PE for women at risk of this condition. Copyright © 2017 ISUOG. Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- A. Perales
- Hospital Universitario y Politécnico La FeValenciaSpain
| | | | | | | | | | | | | | - B. Laíz
- Hospital Universitario y Politécnico La FeValenciaSpain
| | - M. Duque
- Hospital Universitario La PazMadridSpain
| | - M. Navarro
- Hospital Universitario Materno Infantil de CanariasGran CanariaSpain
| | - P. Calmarza
- Hospital Universitario Miguel ServetZaragozaSpain
| | - M. Hund
- Roche Diagnostics International LtdRotkreuzSwitzerland
| | - F. V. Álvarez
- Hospital Universitario Central de AsturiasOviedoSpain
| |
Collapse
|
24
|
Soni S, Krantz DA, Greenberg M, Vohra N, Rochelson B. Recurrence of extreme serum analytes in consecutive pregnancies and association with obstetrical outcomes . J Matern Fetal Neonatal Med 2017; 32:85-91. [PMID: 28838266 DOI: 10.1080/14767058.2017.1371695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVE To evaluate if presence of extreme maternal serum biochemical analytes recurs in consecutive pregnancies. We hypothesized that presence of >1 extreme analyte in prior pregnancy is associated with increased risk of adverse pregnancy outcome in subsequent pregnancy. METHODS Retrospective cohort study of singleton pregnancies evaluated and delivered in 2 consecutive pregnancies (2011-2015). Adverse outcomes were defined as indicated preterm delivery before 37 completed weeks due to preeclampsia, fetal growth restriction or other complications. RESULTS First and second trimester maternal serum analytes were assessed in 1434 patients in 2 consecutive pregnancies. The presence of >1 extreme serum analyte in prior pregnancy significantly increased likelihood of >1 extreme analyte in subsequent pregnancy. The likelihood increased as number of prior extreme markers increased. In patients with normal outcomes and 2 or more extreme serum analytes in prior pregnancy, there was an increased incidence of adverse pregnancy outcomes in subsequent pregnancy with relative risk (RR) of 5.42 [95% CI 1.6-18.3]. CONCLUSIONS The presence of more than 1 extreme serum marker in one pregnancy increases likelihood of recurrence in subsequent pregnancies. Risk of adverse outcomes in subsequent pregnancy can be evaluated based on biochemistry results as well as prior pregnancy outcomes.
Collapse
Affiliation(s)
- Shelly Soni
- a Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology , Hofstra-Northwell School of Medicine , Manhasset , NY , USA
| | | | - Meir Greenberg
- c Division of Obgyn Medical Informatics, Department of Obstetrics and Gynecology , Hofstra-Northwell School of Medicine , Manhasset , NY , USA
| | - Nidhi Vohra
- a Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology , Hofstra-Northwell School of Medicine , Manhasset , NY , USA
| | - Burton Rochelson
- a Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology , Hofstra-Northwell School of Medicine , Manhasset , NY , USA
| |
Collapse
|
25
|
Oudejans CB, Poutsma A, Michel OJ, Thulluru HK, Mulders J, van de Vrugt HJ, Sistermans EA, van Dijk M. Noncoding RNA-regulated gain-of-function of STOX2 in Finnish pre-eclamptic families. Sci Rep 2016; 6:32129. [PMID: 27555360 PMCID: PMC4995371 DOI: 10.1038/srep32129] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/27/2016] [Indexed: 11/09/2022] Open
Abstract
The familial forms of early onset pre-eclampsia and related syndromes (HELLP) present with hypertension and proteinuria in the mother and growth restriction of the fetus. Genetically, these clinically similar entities are caused by different founder-dependent, placentally-expressed paralogous genes. All susceptibility genes (STOX1, lincHELLP, INO80B) identified so far are master control genes that regulate an essential trophoblast differentiation pathway, but act at different entry points. Many genes remain to be identified. Here we demonstrate that a long non-coding RNA (lncRNA) within intron 3 of the STOX2 gene on 4q35.1 acts as a permissive cis-acting regulator of alternative splicing of STOX2. When this lncRNA is mutated or absent, an alternative exon (3B) of STOX2 is included. This introduces a stop codon resulting in the deletion of a highly conserved domain of 64 amino acids in the C-terminal of the STOX2 protein. A mutation present within a regulatory region within intron 1 of STOX2 has the same effect after blocking with CRISPR technology: transcripts with exon 3B are upregulated. This proces appears related to transcriptional control by a chromatin-splicing adaptor complex as described for FGFR2. For STOX2, CHD5, coding for a chromodomain helicase DNA binding protein, qualifies as the chromatin modifier in this process.
Collapse
Affiliation(s)
- Cees Bm Oudejans
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, the Netherlands
| | - Ankie Poutsma
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, the Netherlands
| | - Omar J Michel
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, the Netherlands
| | - Hari K Thulluru
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, the Netherlands
| | - Joyce Mulders
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, the Netherlands
| | - Henri J van de Vrugt
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Erik A Sistermans
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Marie van Dijk
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
26
|
Wan JP, Li L, Li HY, Wang F, Zhang XJ, Zhao H, Li CZ, Wang XT, Chen ZJ. Role of UMOD Promoter Polymorphism in the Etiology of Preeclampsia. Genet Test Mol Biomarkers 2016; 20:471-4. [PMID: 27315129 DOI: 10.1089/gtmb.2015.0268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Ji-Peng Wan
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Lei Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Hong-Yan Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Fei Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Xiao-Jing Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Han Zhao
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key laboratory for Reproductive Endocrinology of Ministry of Education, Jinan, China
- Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China
| | - Chang-Zhong Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Xie-Tong Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key laboratory for Reproductive Endocrinology of Ministry of Education, Jinan, China
- Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China
| |
Collapse
|
27
|
Lekva T, Lyle R, Roland MCP, Friis C, Bianchi DW, Jaffe IZ, Norwitz ER, Bollerslev J, Henriksen T, Ueland T. Gene expression in term placentas is regulated more by spinal or epidural anesthesia than by late-onset preeclampsia or gestational diabetes mellitus. Sci Rep 2016; 6:29715. [PMID: 27405415 PMCID: PMC4942618 DOI: 10.1038/srep29715] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/22/2016] [Indexed: 11/10/2022] Open
Abstract
Pre-eclampsia (PE) and gestational diabetes mellitus (GDM) are common complications of pregnancy, but the mechanisms underlying these disorders remain unclear. The aim was to identify the extent of altered gene expression in term placentas from pregnant women with late-onset PE and GDM compared to controls. RNAseq identified few significantly differentially regulated genes in placental biopsies between PE, GDM, or uncomplicated pregnancy (n = 10 each group). Five genes were altered in placentas from PE including 4 non-coding genes and Angiopoietin 2 (ANGPT2). No genes were significantly regulated by GDM. In contrast, many genes were significantly regulated by fetal, maternal and delivery-specific variables, particularly spinal and epidural anesthesia. We selected ANGPT2 and Chemokine (C-X-C motif) ligand 14 (CXCL14) to test with qPCR in a larger set of placentas (n = 475) and found no differences between the groups. However, regression analysis revealed a stronger association between placental ANGPT2 and CXCL14 mRNA expression and fetal, maternal and delivery-specific variables than diagnostic group. To conclude, the gene expression in term placentas are highly affected by fetal, maternal and delivery specific variables. Few regulated genes were found in late-onset PE and GDM placentas, which may suggest that these conditions could be more affected by maternal factors.
Collapse
Affiliation(s)
- Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Mother Infant Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Robert Lyle
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | | | - Camilla Friis
- Department of Obstetrics, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Diana W Bianchi
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute and Division of Cardiology, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Errol R Norwitz
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA, USA.,Department of Obstetrics &Gynecology, Tufts Medical Center and Tufts University School of Medicine, Boston, MA, USA
| | - Jens Bollerslev
- Faculty of Medicine, University of Oslo, Oslo, Norway.,Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Tore Henriksen
- Department of Obstetrics, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
28
|
White WM, Sun Z, Borowski KS, Brost BC, Davies NP, Rose CH, Garovic VD. Preeclampsia/Eclampsia candidate genes show altered methylation in maternal leukocytes of preeclamptic women at the time of delivery. Hypertens Pregnancy 2016; 35:394-404. [PMID: 27064514 DOI: 10.3109/10641955.2016.1162315] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To analyze methylation profiles of known preeclampsia/eclampsia (PE) candidate genes in normal (NL) and preeclamptic (PE) women at delivery. METHODS A matched case-control study comparing methylation in 79 CpG sites/33 genes from an independent gene set in maternal leukocyte DNA in PE and NL (n = 14 each) on an Illumina BeadChip platform. Replication performed on second cohort (PE = 12; NL = 32). RESULTS PE demonstrates differential methylation in POMC, AGT, CALCA, and DDAH1 compared with NL. CONCLUSION Differential methylation in four genes associated with PE may represent a potential biomarker or an epigenetic pathophysiologic mechanism altering gene transcription.
Collapse
Affiliation(s)
- Wendy M White
- a Department of Obstetrics and Gynecology , Division of Maternal Fetal Medicine , Mayo Clinic, Rochester , MN , USA
| | - Zhifu Sun
- b Department of Health Sciences Research , Division of Biomedical Statistics and Informatics , Mayo Clinic, Rochester , MN , USA
| | - Kristi S Borowski
- a Department of Obstetrics and Gynecology , Division of Maternal Fetal Medicine , Mayo Clinic, Rochester , MN , USA
| | - Brian C Brost
- a Department of Obstetrics and Gynecology , Division of Maternal Fetal Medicine , Mayo Clinic, Rochester , MN , USA
| | - Norman P Davies
- a Department of Obstetrics and Gynecology , Division of Maternal Fetal Medicine , Mayo Clinic, Rochester , MN , USA
| | - Carl H Rose
- a Department of Obstetrics and Gynecology , Division of Maternal Fetal Medicine , Mayo Clinic, Rochester , MN , USA
| | - Vesna D Garovic
- c Department of Internal Medicine , Division of Nephrology and Hypertension , Mayo Clinic, Rochester , MN , USA
| |
Collapse
|
29
|
Kim HJ, Kim SY, Lim JH, Kwak DW, Park SY, Ryu HM. Quantification and Application of Potential Epigenetic Markers in Maternal Plasma of Pregnancies with Hypertensive Disorders. Int J Mol Sci 2015; 16:29875-88. [PMID: 26694356 PMCID: PMC4691144 DOI: 10.3390/ijms161226201] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 12/01/2015] [Accepted: 12/08/2015] [Indexed: 12/18/2022] Open
Abstract
The aim of this study was to evaluate quantitative aberrations of novel fetal-specific epigenetic markers in maternal plasma of pregnancies with hypertensive disorders. We compared the concentrations of DSCR3, RASSF1A, and SRY as cell-free fetal DNA markers in 188 normal pregnancies, 16 pregnancies with early-onset preeclampsia (EO-PE), 47 pregnancies with late-onset preeclampsia (LO-PE), and 29 pregnancies with gestational hypertension (GH). The concentrations of all markers were significantly correlated with gestational age (p < 0.001 for all). Strong positive correlations were also observed between DSCR3 and SRY (r = 0.471, p < 0.001), as well as between RASSF1A and SRY (r = 0.326, p = 0.015) and between DSCR3 and RASSF1A (r = 0.673, p < 0.001). The concentrations of DSCR3 and RASSF1A in the EO-PE were significantly higher at 24–32 weeks and onwards (p < 0.05 for both). In the LO-PE, DSCR3 and RASSF1A concentrations were significantly higher only at 33–41 weeks compared with the controls. The concentrations of all markers in the GH group were not significantly different from those in the control group. This study is the first demonstration that DSCR3 is a novel epigenetic marker that can be an alternative to the RASSF1A for the prediction of EO-PE.
Collapse
Affiliation(s)
- Hyun Jin Kim
- Laboratory of Medical Genetics, Medical Research Institute, Cheil General Hospital and Women's Healthcare Center, Seoul 100-380, Korea.
| | - Shin Young Kim
- Laboratory of Medical Genetics, Medical Research Institute, Cheil General Hospital and Women's Healthcare Center, Seoul 100-380, Korea.
| | - Ji Hyae Lim
- Laboratory of Medical Genetics, Medical Research Institute, Cheil General Hospital and Women's Healthcare Center, Seoul 100-380, Korea.
| | - Dong Wook Kwak
- Department of Obstetrics and Gynecology, Cheil General Hospital and Women's Healthcare Center, Dankook University College of Medicine, Seoul 100-380, Korea.
| | - So Yeon Park
- Laboratory of Medical Genetics, Medical Research Institute, Cheil General Hospital and Women's Healthcare Center, Seoul 100-380, Korea.
| | - Hyun Mee Ryu
- Laboratory of Medical Genetics, Medical Research Institute, Cheil General Hospital and Women's Healthcare Center, Seoul 100-380, Korea.
- Department of Obstetrics and Gynecology, Cheil General Hospital and Women's Healthcare Center, Dankook University College of Medicine, Seoul 100-380, Korea.
| |
Collapse
|
30
|
Oudejans CB. Maternal plasma RNA sequencing. Clin Biochem 2015; 48:942-7. [DOI: 10.1016/j.clinbiochem.2015.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/06/2015] [Accepted: 03/08/2015] [Indexed: 01/08/2023]
|
31
|
Kaartokallio T, Cervera A, Kyllönen A, Laivuori K, Kere J, Laivuori H. Gene expression profiling of pre-eclamptic placentae by RNA sequencing. Sci Rep 2015; 5:14107. [PMID: 26388242 PMCID: PMC4585671 DOI: 10.1038/srep14107] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/24/2015] [Indexed: 01/23/2023] Open
Abstract
Pre-eclampsia is a common and complex pregnancy disorder that often involves impaired placental development. In order to identify altered gene expression in pre-eclamptic placenta, we sequenced placental transcriptomes of nine pre-eclamptic and nine healthy pregnant women in pools of three. The differential gene expression was tested both by including all the pools in the analysis and by excluding some of the pools based on phenotypic characteristics. From these analyses, we identified altogether 53 differently expressed genes, a subset of which was validated by qPCR in 20 cases and 19 controls. Furthermore, we conducted pathway and functional analyses which revealed disturbed vascular function and immunological balance in pre-eclamptic placenta. Some of the genes identified in our study have been reported by numerous microarray studies (BHLHE40, FSTL3, HK2, HTRA4, LEP, PVRL4, SASH1, SIGLEC6), but many have been implicated in only few studies or have not previously been linked to pre-eclampsia (ARMS2, BTNL9, CCSAP, DIO2, FER1L4, HPSE, LOC100129345, LYN, MYO7B, NCMAP, NDRG1, NRIP1, PLIN2, SBSPON, SERPINB9, SH3BP5, TET3, TPBG, ZNF175). Several of the molecules produced by these genes may have a role in the pathogenesis of pre-eclampsia, and some could qualify as biomarkers for prediction or detection of this pregnancy complication.
Collapse
Affiliation(s)
- Tea Kaartokallio
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, FI-00014, Finland
| | - Alejandra Cervera
- Research Programs Unit, Genome-Scale Biology and Institute of Biomedicine, University of Helsinki, Helsinki, FI-00014, Finland
| | - Anjuska Kyllönen
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, FI-00014, Finland
| | - Krista Laivuori
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, FI-00014, Finland.,Molecular Neurology Research Program, University of Helsinki, Helsinki, FI-00014, Finland.,Folkhälsan Institute of Genetics, Helsinki, FI-000290, Finland
| | - Juha Kere
- Molecular Neurology Research Program, University of Helsinki, Helsinki, FI-00014, Finland.,Folkhälsan Institute of Genetics, Helsinki, FI-000290, Finland.,Department of Biosciences and Nutrition, Center for Innovative Medicine, and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Hannele Laivuori
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, FI-00014, Finland.,Obstetrics and Gynaecology, University of Helsinki and Helsinki University Hospital, Helsinki, FI-00014, Finland.,Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, FI-00014, Finland
| | | |
Collapse
|
32
|
Pilliod RA, Feinberg BB, Burwick RM. Maternal and feto-placental phenotypes of early-onset severe preeclampsia. J Matern Fetal Neonatal Med 2015; 29:1209-13. [DOI: 10.3109/14767058.2015.1045867] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
33
|
Pre-eclampsia causes adverse maternal outcomes across the gestational spectrum. Pregnancy Hypertens 2015; 5:198-204. [DOI: 10.1016/j.preghy.2015.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 02/03/2015] [Accepted: 02/14/2015] [Indexed: 11/19/2022]
|
34
|
Liu Y, Zhao Y, Yu A, Zhao B, Gao Y, Niu H. Diagnostic accuracy of the soluble Fms-like tyrosine kinase-1/placental growth factor ratio for preeclampsia: a meta-analysis based on 20 studies. Arch Gynecol Obstet 2015; 292:507-18. [PMID: 25716670 DOI: 10.1007/s00404-015-3671-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 02/16/2015] [Indexed: 12/20/2022]
Abstract
PURPOSE To investigate the accuracy of the soluble fms-like tyrosine kinase-1(sFlt-1)/placental growth factor (PlGF) ratio to predict preeclampsia (PE). METHODS A search in the PubMed, Embase, and Cochrane Library was conducted for human studies without language restriction. We included the studies reported sufficient data to reconstruct the diagnostic 2 × 2 table of sFlt-1/PlGF with testing of PlGF and sFlt-1 in serum or plasma. Two reviewers independently screened the articles. Disagreements were resolved by a third reviewer. Unpublished data were requested from the authors of the study by email. RESULTS Twenty studies with 28 groups of women with different gestational ages were included in our study. The pooled diagnostic sensitivity and specificity of sFlt-1/PlGF were 0.78 and 0.84 with the area under the SROC curve (AUC) of 0.88. In subgroup analyses, the diagnostic value of sFlt-1/PlGF for early-onset PE is highest with a pooled diagnostic odds ratio (DOR) of 241 and AUC of 0.98. CONCLUSIONS The accuracy of sFlt-1/PlGF ratio for screening PE was moderate and was high for early-onset PE. High-quality studies are needed to confirm their usefulness in prediction of PE in clinical practice.
Collapse
Affiliation(s)
- Yuxiu Liu
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | | | | | | | | | | |
Collapse
|
35
|
Population-based biomarker screening and the development of severe preeclampsia in California. Am J Obstet Gynecol 2014; 211:377.e1-8. [PMID: 24631701 DOI: 10.1016/j.ajog.2014.03.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 02/06/2014] [Accepted: 03/10/2014] [Indexed: 11/20/2022]
Abstract
OBJECTIVE The purpose of this study was to examine the relationship between second-trimester maternal serum biomarkers and the development of early- and late-onset severe preeclampsia in euploid pregnancies. STUDY DESIGN Included were 136,139 pregnancies that obtained second-trimester prenatal screening through the California Prenatal Screening Program with live births in 2006-2008. We identified severe preeclampsia diagnoses from hospital discharge records. We used log binomial regression to examine the association between abnormal second-trimester maternal serum biomarkers and the development of severe preeclampsia. RESULTS Approximately 0.9% of all women (n = 1208) in our sample experienced severe preeclampsia; 329 women at <34 weeks' gestation and 879 women ≥34 weeks' gestation. High levels of alpha fetoprotein (AFP), human chorionic gonadotropin, inhibin (multiple of the median, ≥95th percentile), and low unconjugated estriol (multiple of the median, ≤5th percentile), were associated with severe preeclampsia (relative risk, 2.5-11.7). Biomarkers were more predictive of early-onset severe preeclampsia (relative risk, 3.8-11.7). One in 9.5 pregnancies with combined high AFP, inhibin, and low unconjugated estriol levels experienced severe early-onset preeclampsia compared with 1 in 680.5 pregnancies without any abnormal biomarkers. CONCLUSION The risk of the development of severe preeclampsia increases for women with high second-trimester AFP, human chorionic gonadotropin, inhibin, and/or low unconjugated estriol; this is especially true for early-onset severe preeclampsia. When abnormal biomarkers co-occur, risk dramatically increases. Although the screening value of second-trimester biomarkers is low, abnormal biomarkers, especially when occurring in combination, appear to indicate placental dysfunction that is associated with the development of severe preeclampsia.
Collapse
|
36
|
Yang Y, Liu X, Jia J, Bai Y, Dai L, Wang T, Zhou B, Zhang L, Zhou R. Role of osteoprotegerin gene variants in early-onset severe pre-eclampsia. J Obstet Gynaecol Res 2014; 41:334-42. [PMID: 25256810 DOI: 10.1111/jog.12533] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 07/06/2014] [Indexed: 11/29/2022]
Abstract
AIM The aim of this study was to detect the role of osteoprotegerin (OPG) gene variants in early-onset severe pre-eclampsia. MATERIAL AND METHODS The associations of 163A/G (rs3102735) and 950T/C (rs2073617) polymorphisms of OPG with pre-eclampsia (60 cases of early-onset severe pre-eclampsia and 91 cases of late-onset pre-eclampsia), as well as with the clinical manifestations of individuals, were evaluated. RESULTS Data showed lower frequencies of TC and TC + CC genotypes and C allele of 950T/C in the early-onset group than those of the control and late-onset groups (P = 0.003; P = 0.002; P = 0.005; P = 0.031; P = 0.021; P = 0.022). However, no significant differences were found in genotype and allele frequencies between the late-onset and control groups. Moreover, no significant differences were observed in the genotype and allele frequencies of 163A/G polymorphism among the three groups. In the early-onset group, 950T/C TC + CC genotype carriers exhibited significantly lower systolic blood pressure ([147.25 ± 11.89] mmHg) and 24-h urine protein ([2.46 ± 0.92] g) than the TT carriers ([165.88 ± 20.39] mmHg, [3.64 ± 0.81] g) (P = 0.003; P = 0.001, respectively). Serum creatinine was significantly higher in women with the 163A/G AG + GG genotypes ([82.31 ± 11.66] μmol/L) than in those with the AA genotype ([71.90 ± 16.85] μmol/L) (P = 0.003). CONCLUSION This study implicates that OPG gene variants may be associated with early-onset severe pre-eclampsia.
Collapse
Affiliation(s)
- Yan Yang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Qiao C, Wang C, Jin F, Zheng D, Liu C. Expression of collapsin response mediator protein 1 in placenta of normal gestation and link to early-onset preeclampsia. Reprod Sci 2014; 22:495-501. [PMID: 25194153 DOI: 10.1177/1933719114549847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
A human isoform of Collapsin Response Mediator Protein (CRMP) family proteins, CRMP-1, has been identified as a novel invasion suppressor. The aim of this study was to determine CRMP-1 expression pattern in placentas during normal pregnancy and elucidate the clinical significance of CRMP-1 expression in the placentas of women with early-onset preeclamptic pregnancies. We recruited 66 normal healthy pregnant Chinese women and 60 Chinese patients with preeclampsia [early-onset prereclampsia(ePE), n = 30 and late-onset preeclampsia(lPE) n = 30]. Gestational age-matched normal healthy pregnant women were used as controls of early-onset and late-onset preeclampsia, which were 23-33 + 6 weeks, n = 18 and control B: 34-40 weeks, n = 20). Quantitative RT-PCR, Western blot analysis and immunohistochemistry were used to analyze the expressions of CRMP-1 in placentas. Expression of CRMP-1 was detected in syncytio- and cytotrophoblasts of all groups using immunohistochemistry. CRMP-1 was most abundantly expressed in syncytiotrophoblasts, moderately in cytotrophoblasts and the intermediate trophoblasts especially in the first trimester. The placental expression of CRMP-1 is particularly striking in the first trimester and decreases throughout pregnancy. There is a significant increase in CRMP-1 expression in the placenta of ePE but not of lPE, as compared to gestational-matched controls. The aberrant upregulation of CRMP-1 expression may link to the mechanism of developing ePE.
Collapse
Affiliation(s)
- Chong Qiao
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Chunhui Wang
- Department of Hepatobiliary Surgery, General Hospital of Shenyang Military Region, Shenyang, China
| | - Feng Jin
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Dongying Zheng
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Caixia Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, China
| |
Collapse
|
38
|
Yang P, Dai A, Alexenko AP, Liu Y, Stephens AJ, Schulz LC, Schust DJ, Roberts RM, Ezashi T. Abnormal oxidative stress responses in fibroblasts from preeclampsia infants. PLoS One 2014; 9:e103110. [PMID: 25058409 PMCID: PMC4110005 DOI: 10.1371/journal.pone.0103110] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 06/27/2014] [Indexed: 01/17/2023] Open
Abstract
Background Signs of severe oxidative stress are evident in term placentae of infants born to mothers with preeclampsia (PE), but it is unclear whether this is a cause or consequence of the disease. Here fibroblast lines were established from umbilical cords (UC) delivered by mothers who had experienced early onset PE and from controls with the goal of converting these primary cells to induced pluripotent stem cells and ultimately trophoblast. Contrary to expectations, the oxidative stress responses of these non-placental cells from PE infants were more severe than those from controls. Methods and Findings Three features suggested that UC-derived fibroblasts from PE infants responded less well to oxidative stressors than controls: 1) While all UC provided outgrowths in 4% O2, success was significantly lower for PE cords in 20% O2; 2) PE lines established in 4% O2 proliferated more slowly than controls when switched to 20% O2; 3) PE lines were more susceptible to the pro-oxidants diethylmaleate and tert-butylhydroquinone than control lines, but, unlike controls, were not protected by glutathione. Transcriptome profiling revealed only a few genes differentially regulated between PE lines and controls in 4% O2 conditions. However, a more severely stressed phenotype than controls, particularly in the unfolded protein response, was evident when PE lines were switched suddenly to 20% O2, thus confirming the greater sensitivity of the PE fibroblasts to acute changes in oxidative stress. Conclusions UC fibroblasts derived from PE infants are intrinsically less able to respond to acute oxidative stress than controls, and this phenotype is retained over many cell doublings. Whether the basis of this vulnerability is genetic or epigenetic and how it pertains to trophoblast development remains unclear, but this finding may provide a clue to the basis of the early onset, usually severe, form of PE.
Collapse
Affiliation(s)
- Penghua Yang
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, United States of America
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
- College of Life Science, Yangtze University, Jingzhou, Hubei, China
| | - Aihua Dai
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, United States of America
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Andrei P. Alexenko
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, United States of America
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Yajun Liu
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, United States of America
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
- Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Amanda J. Stephens
- Department of Obstetrics, Gynecology, & Women’s Health, University of Missouri, Columbia, Missouri, United States of America
| | - Laura C. Schulz
- Department of Obstetrics, Gynecology, & Women’s Health, University of Missouri, Columbia, Missouri, United States of America
| | - Danny J. Schust
- Department of Obstetrics, Gynecology, & Women’s Health, University of Missouri, Columbia, Missouri, United States of America
| | - R. Michael Roberts
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, United States of America
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
| | - Toshihiko Ezashi
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, United States of America
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
39
|
Genetic aspects of preeclampsia and the HELLP syndrome. J Pregnancy 2014; 2014:910751. [PMID: 24991435 PMCID: PMC4060423 DOI: 10.1155/2014/910751] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 02/27/2014] [Accepted: 04/01/2014] [Indexed: 12/20/2022] Open
Abstract
Both preeclampsia and the HELLP syndrome have their origin in the placenta. The aim of this study is to review genetic factors involved in development of preeclampsia and the HELLP syndrome using literature search in PubMed. A familial cohort links chromosomes 2q, 5q, and 13q to preeclampsia. The chromosome 12q is coupled with the HELLP syndrome. The STOX1 gene, the ERAP1 and 2 genes, the syncytin envelope gene, and the −670 Fas receptor polymorphisms are involved in the development of preeclampsia. The ACVR2A gene on chromosome 2q22 is also implicated. The toll-like receptor-4 (TLR-4) and factor V Leiden mutation participate both in development of preeclampsia and the HELLP syndrome. Carriers of the TT and the CC genotype of the MTHFR C677T polymorphism seem to have an increased risk of the HELLP syndrome. The placental levels of VEGF mRNA are reduced both in women with preeclampsia and in women with the HELLP syndrome. The BclI polymorphism is engaged in development of the HELLP syndrome but not in development of severe preeclampsia. The ACE I/D polymorphism affects uteroplacental and umbilical artery blood flows in women with preeclampsia. In women with preeclampsia and the HELLP syndrome several genes in the placenta are deregulated. Preeclampsia and the HELLP syndrome are multiplex genetic diseases.
Collapse
|
40
|
Kaartokallio T, Klemetti MM, Timonen A, Uotila J, Heinonen S, Kajantie E, Kere J, Kivinen K, Pouta A, Lakkisto P, Laivuori H. Microsatellite polymorphism in the heme oxygenase-1 promoter is associated with nonsevere and late-onset preeclampsia. Hypertension 2014; 64:172-7. [PMID: 24799610 DOI: 10.1161/hypertensionaha.114.03337] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Preeclampsia is a serious and phenotypically heterogeneous vascular pregnancy disorder. Heme oxygenase-1 (HO-1) is a stress response enzyme that may protect the maternal endothelium and facilitate adequate metabolic adaptation to pregnancy by its antioxidant and anti-inflammatory functions. HO-1 stress response is modulated by HO-1 gene (HMOX1) polymorphisms. Individuals with the long allele of a guanine-thymine (GTn) microsatellite repeat located in the promoter region of HMOX1 have a higher risk of cardiometabolic diseases compared with those with the short allele. We investigated whether the long GTn allele of HMOX1 is associated with subtypes of preeclampsia. The GTn repeat was genotyped in 759 patients and in 779 controls from the Finnish Genetics of Preeclampsia Consortium (FINNPEC) cohort using DNA fragment analysis. In subtype analyses, the long-long (LL) genotype was associated with nonsevere (additive model: odds ratio [OR], 1.94; 95% confidence interval [CI], 1.13-3.31; recessive model: OR, 1.39; 95% CI, 1.02-1.89) and late-onset (additive model: OR, 1.44; 95% CI, 1.02-2.05; recessive model: OR, 1.28; 95% CI, 1.02-1.59) preeclampsia and with preeclampsia without a small-for-gestational-age infant (recessive model: OR, 1.27; 95% CI, 1.02-1.58). The long allele was associated with nonsevere (OR, 1.35; 95% CI, 1.07-1.70) and late-onset (OR, 1.21; 95% CI, 1.03-1.42) preeclampsia and with preeclampsia without a small-for-gestational-age infant (OR, 1.19; 95% CI, 1.02-1.40). Moreover, both the LL genotype and the long allele were associated with preeclampsia in women who had smoked during pregnancy. In conclusion, the GTn long allele seems to predispose to late-onset, less severe form of preeclampsia. This finding supports the role of HO-1 in the pathogenesis of preeclampsia and suggests that the HO-1 pathway may provide a potential target for the treatment of preeclampsia.
Collapse
Affiliation(s)
- Tea Kaartokallio
- From the Haartman Institute, Medical Genetics (T.K., M.M.K., A.T., J.K., H.L.), Research Programs Unit, Molecular Neurology (A.T.), Department of Clinical Chemistry (P.L.), and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, Helsinki, Finland; Departments of Obstetrics and Gynecology (M.M.K., H.L.) and Clinical Chemistry (P.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Obstetrics and Gynecology, South-Karelia Central Hospital, Lappeenranta, Finland (M.M.K.); Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland (J.U.); Department of Obstetrics and Gynecology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland (S.H.); Department of Chronic Disease Prevention, Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland (E.K.); Department of Children, Young People, and Families, National Institute for Health and Welfare, Oulu, Finland (A.P.); Children's Hospital, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland (E.K.); Department of Obstetrics and Gynaecology, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland (E.K., A.P.); Department of Biosciences and Nutrition, and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden (J.K.); Folkhälsan Institute of Genetics, Helsinki, Finland (J.K.); Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (K.K.); and Minerva Institute for Medical Research, Helsinki, Finland (P.L.).
| | - Miira M Klemetti
- From the Haartman Institute, Medical Genetics (T.K., M.M.K., A.T., J.K., H.L.), Research Programs Unit, Molecular Neurology (A.T.), Department of Clinical Chemistry (P.L.), and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, Helsinki, Finland; Departments of Obstetrics and Gynecology (M.M.K., H.L.) and Clinical Chemistry (P.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Obstetrics and Gynecology, South-Karelia Central Hospital, Lappeenranta, Finland (M.M.K.); Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland (J.U.); Department of Obstetrics and Gynecology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland (S.H.); Department of Chronic Disease Prevention, Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland (E.K.); Department of Children, Young People, and Families, National Institute for Health and Welfare, Oulu, Finland (A.P.); Children's Hospital, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland (E.K.); Department of Obstetrics and Gynaecology, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland (E.K., A.P.); Department of Biosciences and Nutrition, and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden (J.K.); Folkhälsan Institute of Genetics, Helsinki, Finland (J.K.); Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (K.K.); and Minerva Institute for Medical Research, Helsinki, Finland (P.L.)
| | - Anni Timonen
- From the Haartman Institute, Medical Genetics (T.K., M.M.K., A.T., J.K., H.L.), Research Programs Unit, Molecular Neurology (A.T.), Department of Clinical Chemistry (P.L.), and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, Helsinki, Finland; Departments of Obstetrics and Gynecology (M.M.K., H.L.) and Clinical Chemistry (P.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Obstetrics and Gynecology, South-Karelia Central Hospital, Lappeenranta, Finland (M.M.K.); Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland (J.U.); Department of Obstetrics and Gynecology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland (S.H.); Department of Chronic Disease Prevention, Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland (E.K.); Department of Children, Young People, and Families, National Institute for Health and Welfare, Oulu, Finland (A.P.); Children's Hospital, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland (E.K.); Department of Obstetrics and Gynaecology, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland (E.K., A.P.); Department of Biosciences and Nutrition, and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden (J.K.); Folkhälsan Institute of Genetics, Helsinki, Finland (J.K.); Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (K.K.); and Minerva Institute for Medical Research, Helsinki, Finland (P.L.)
| | - Jukka Uotila
- From the Haartman Institute, Medical Genetics (T.K., M.M.K., A.T., J.K., H.L.), Research Programs Unit, Molecular Neurology (A.T.), Department of Clinical Chemistry (P.L.), and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, Helsinki, Finland; Departments of Obstetrics and Gynecology (M.M.K., H.L.) and Clinical Chemistry (P.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Obstetrics and Gynecology, South-Karelia Central Hospital, Lappeenranta, Finland (M.M.K.); Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland (J.U.); Department of Obstetrics and Gynecology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland (S.H.); Department of Chronic Disease Prevention, Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland (E.K.); Department of Children, Young People, and Families, National Institute for Health and Welfare, Oulu, Finland (A.P.); Children's Hospital, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland (E.K.); Department of Obstetrics and Gynaecology, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland (E.K., A.P.); Department of Biosciences and Nutrition, and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden (J.K.); Folkhälsan Institute of Genetics, Helsinki, Finland (J.K.); Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (K.K.); and Minerva Institute for Medical Research, Helsinki, Finland (P.L.)
| | - Seppo Heinonen
- From the Haartman Institute, Medical Genetics (T.K., M.M.K., A.T., J.K., H.L.), Research Programs Unit, Molecular Neurology (A.T.), Department of Clinical Chemistry (P.L.), and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, Helsinki, Finland; Departments of Obstetrics and Gynecology (M.M.K., H.L.) and Clinical Chemistry (P.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Obstetrics and Gynecology, South-Karelia Central Hospital, Lappeenranta, Finland (M.M.K.); Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland (J.U.); Department of Obstetrics and Gynecology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland (S.H.); Department of Chronic Disease Prevention, Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland (E.K.); Department of Children, Young People, and Families, National Institute for Health and Welfare, Oulu, Finland (A.P.); Children's Hospital, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland (E.K.); Department of Obstetrics and Gynaecology, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland (E.K., A.P.); Department of Biosciences and Nutrition, and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden (J.K.); Folkhälsan Institute of Genetics, Helsinki, Finland (J.K.); Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (K.K.); and Minerva Institute for Medical Research, Helsinki, Finland (P.L.)
| | - Eero Kajantie
- From the Haartman Institute, Medical Genetics (T.K., M.M.K., A.T., J.K., H.L.), Research Programs Unit, Molecular Neurology (A.T.), Department of Clinical Chemistry (P.L.), and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, Helsinki, Finland; Departments of Obstetrics and Gynecology (M.M.K., H.L.) and Clinical Chemistry (P.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Obstetrics and Gynecology, South-Karelia Central Hospital, Lappeenranta, Finland (M.M.K.); Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland (J.U.); Department of Obstetrics and Gynecology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland (S.H.); Department of Chronic Disease Prevention, Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland (E.K.); Department of Children, Young People, and Families, National Institute for Health and Welfare, Oulu, Finland (A.P.); Children's Hospital, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland (E.K.); Department of Obstetrics and Gynaecology, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland (E.K., A.P.); Department of Biosciences and Nutrition, and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden (J.K.); Folkhälsan Institute of Genetics, Helsinki, Finland (J.K.); Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (K.K.); and Minerva Institute for Medical Research, Helsinki, Finland (P.L.)
| | - Juha Kere
- From the Haartman Institute, Medical Genetics (T.K., M.M.K., A.T., J.K., H.L.), Research Programs Unit, Molecular Neurology (A.T.), Department of Clinical Chemistry (P.L.), and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, Helsinki, Finland; Departments of Obstetrics and Gynecology (M.M.K., H.L.) and Clinical Chemistry (P.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Obstetrics and Gynecology, South-Karelia Central Hospital, Lappeenranta, Finland (M.M.K.); Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland (J.U.); Department of Obstetrics and Gynecology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland (S.H.); Department of Chronic Disease Prevention, Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland (E.K.); Department of Children, Young People, and Families, National Institute for Health and Welfare, Oulu, Finland (A.P.); Children's Hospital, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland (E.K.); Department of Obstetrics and Gynaecology, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland (E.K., A.P.); Department of Biosciences and Nutrition, and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden (J.K.); Folkhälsan Institute of Genetics, Helsinki, Finland (J.K.); Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (K.K.); and Minerva Institute for Medical Research, Helsinki, Finland (P.L.)
| | - Katja Kivinen
- From the Haartman Institute, Medical Genetics (T.K., M.M.K., A.T., J.K., H.L.), Research Programs Unit, Molecular Neurology (A.T.), Department of Clinical Chemistry (P.L.), and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, Helsinki, Finland; Departments of Obstetrics and Gynecology (M.M.K., H.L.) and Clinical Chemistry (P.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Obstetrics and Gynecology, South-Karelia Central Hospital, Lappeenranta, Finland (M.M.K.); Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland (J.U.); Department of Obstetrics and Gynecology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland (S.H.); Department of Chronic Disease Prevention, Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland (E.K.); Department of Children, Young People, and Families, National Institute for Health and Welfare, Oulu, Finland (A.P.); Children's Hospital, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland (E.K.); Department of Obstetrics and Gynaecology, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland (E.K., A.P.); Department of Biosciences and Nutrition, and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden (J.K.); Folkhälsan Institute of Genetics, Helsinki, Finland (J.K.); Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (K.K.); and Minerva Institute for Medical Research, Helsinki, Finland (P.L.)
| | - Anneli Pouta
- From the Haartman Institute, Medical Genetics (T.K., M.M.K., A.T., J.K., H.L.), Research Programs Unit, Molecular Neurology (A.T.), Department of Clinical Chemistry (P.L.), and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, Helsinki, Finland; Departments of Obstetrics and Gynecology (M.M.K., H.L.) and Clinical Chemistry (P.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Obstetrics and Gynecology, South-Karelia Central Hospital, Lappeenranta, Finland (M.M.K.); Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland (J.U.); Department of Obstetrics and Gynecology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland (S.H.); Department of Chronic Disease Prevention, Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland (E.K.); Department of Children, Young People, and Families, National Institute for Health and Welfare, Oulu, Finland (A.P.); Children's Hospital, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland (E.K.); Department of Obstetrics and Gynaecology, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland (E.K., A.P.); Department of Biosciences and Nutrition, and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden (J.K.); Folkhälsan Institute of Genetics, Helsinki, Finland (J.K.); Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (K.K.); and Minerva Institute for Medical Research, Helsinki, Finland (P.L.)
| | - Päivi Lakkisto
- From the Haartman Institute, Medical Genetics (T.K., M.M.K., A.T., J.K., H.L.), Research Programs Unit, Molecular Neurology (A.T.), Department of Clinical Chemistry (P.L.), and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, Helsinki, Finland; Departments of Obstetrics and Gynecology (M.M.K., H.L.) and Clinical Chemistry (P.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Obstetrics and Gynecology, South-Karelia Central Hospital, Lappeenranta, Finland (M.M.K.); Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland (J.U.); Department of Obstetrics and Gynecology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland (S.H.); Department of Chronic Disease Prevention, Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland (E.K.); Department of Children, Young People, and Families, National Institute for Health and Welfare, Oulu, Finland (A.P.); Children's Hospital, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland (E.K.); Department of Obstetrics and Gynaecology, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland (E.K., A.P.); Department of Biosciences and Nutrition, and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden (J.K.); Folkhälsan Institute of Genetics, Helsinki, Finland (J.K.); Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (K.K.); and Minerva Institute for Medical Research, Helsinki, Finland (P.L.)
| | - Hannele Laivuori
- From the Haartman Institute, Medical Genetics (T.K., M.M.K., A.T., J.K., H.L.), Research Programs Unit, Molecular Neurology (A.T.), Department of Clinical Chemistry (P.L.), and Institute for Molecular Medicine Finland (H.L.), University of Helsinki, Helsinki, Finland; Departments of Obstetrics and Gynecology (M.M.K., H.L.) and Clinical Chemistry (P.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Obstetrics and Gynecology, South-Karelia Central Hospital, Lappeenranta, Finland (M.M.K.); Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland (J.U.); Department of Obstetrics and Gynecology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland (S.H.); Department of Chronic Disease Prevention, Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland (E.K.); Department of Children, Young People, and Families, National Institute for Health and Welfare, Oulu, Finland (A.P.); Children's Hospital, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland (E.K.); Department of Obstetrics and Gynaecology, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland (E.K., A.P.); Department of Biosciences and Nutrition, and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden (J.K.); Folkhälsan Institute of Genetics, Helsinki, Finland (J.K.); Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (K.K.); and Minerva Institute for Medical Research, Helsinki, Finland (P.L.)
| |
Collapse
|
41
|
Procopciuc LM, Zaharie G, Caracostea G, Stamatian F. Newborn LpL (Ser447Stop, Asn291Ser) genotypes and the interaction with maternal genotypes influence the risk for different types of preeclampsia: modulating effect on lipid profile and pregnancy outcome. Gynecol Endocrinol 2014; 30:221-5. [PMID: 24400599 DOI: 10.3109/09513590.2013.871512] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
AIM To establish that newborn Ser447Stop and Asn291Ser may have interactive effects with maternal genotypes on the plasma lipoprotein levels, risk of preeclampsia as well as on the prognosis of preeclampsia. MATERIALS AND METHODS Seventy preeclamptic women and 94 normotensive pregnant women, and their newborns were genotyped using PCR-RFLP methods. RESULTS The risk of mild and severe preeclampsia was 4 (p = 0.004) and 5.18 (p = 0.001), respectively, if both the mother and newborn were carriers of the Ser447/Ser477 genotype. If both the mother and newborn were carriers of the Asn291Ser variant, the risk to develop severe preeclampsia was 6.07 (p = 0.03). Women with mild and severe preeclampsia had higher TG (p < 0.001; p < 0.001) and LDL-C levels (p = 0.008; p < 0.001) if both the mother and newborn were carriers of the Ser447/Ser447 genotype. Women with severe preeclampsia had significantly higher TG (p = 0.03) and LDL-C levels (p = 0.037) if both the mother and newborn were carriers of Asn291Ser. Newborn/maternal LpL interaction had no statistically significant influence on pregnancy outcome. CONCLUSIONS The newborn/maternal LpL interaction influences the severity of preeclampsia and modulates the lipid profile particularly in severe preeclampsia.
Collapse
|
42
|
Martínez-Orgado J, Bonet B, Sanchez-Vera I. Consequences of preeclampsia for the newborn: role of oxidative stress. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/17474108.2.5.651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
43
|
Wang F, Wang L, Shi Z, Liang G. Comparative N-glycoproteomic and phosphoproteomic profiling of human placental plasma membrane between normal and preeclampsia pregnancies with high-resolution mass spectrometry. PLoS One 2013; 8:e80480. [PMID: 24260401 PMCID: PMC3829899 DOI: 10.1371/journal.pone.0080480] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 10/03/2013] [Indexed: 11/22/2022] Open
Abstract
Preeclampsia is a serious complication of pregnancy, which affects 2–8% of all pregnancies and is one of the leading causes of maternal and perinatal mortality and morbidity worldwide. To better understand the molecular mechanisms involved in pathological development of placenta in preeclampsia, we used high-resolution LC-MS/MS technologies to construct a comparative N-glycoproteomic and phosphoproteomic profiling of human placental plasma membrane in normal and preeclamptic pregnancies. A total of 1027 N-glyco- and 2094 phospho- sites were detected in human placental plasma membrane, and 5 N-glyco- and 38 phospho- proteins, respectively, with differentially expression were definitively identified between control and preeclamptic placental plasma membrane. Further bioinformatics analysis indicated that these differentially expressed proteins correlate with several specific cellular processes occurring during pathological changes of preeclamptic placental plasma membrane.
Collapse
Affiliation(s)
- Fuqiang Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Chemistry, University of Science and Technology of China, Anhui, China ; State Key Laboratory of Reproductive Medicine, Analysis Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | | | | | | |
Collapse
|
44
|
Wan JP, Zhao H, Li T, Li CZ, Wang XT, Chen ZJ. The common variant rs11646213 is associated with preeclampsia in Han Chinese women. PLoS One 2013; 8:e71202. [PMID: 23976997 PMCID: PMC3747203 DOI: 10.1371/journal.pone.0071202] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 07/03/2013] [Indexed: 12/21/2022] Open
Abstract
Background Preeclampsia, characterized by hypertension and proteinuria, is a multifactorial disease caused by complex interactions between environmental and genetic factors. A recent genome-wide association study of blood pressure reported an association between hypertension and rs11646213. This study evaluated the association between preeclampsia and rs11646213. Methods A total of 454 cases and 460 controls were recruited to participate in this study. The single nucleotide polymorphism (SNP) rs11646213 was genotyped by polymerase chain reaction (PCR) and direct sequencing. Results The allele frequency of rs11646213 was significantly different between the preeclampsia and control groups (P = 0.017, OR = 1.36, 95% CI = 1.06–1.76). Differences were particularly significant in the severe preeclampsia subgroup (P = 0.002, OR = 1.54, 95% CI = 1.17–2.03) and the early-onset preeclampsia subgroup (P = 0.004, OR = 1.57, 95% CI = 1.16–2.13). Genotyping analysis showed that the T allele of rs11646213 could confer a risk for preeclampsia, severe preeclampsia and early-onset preeclampsia. Conclusions Rs11646213 upstream of the CDH13 gene is associated with preeclampsia in Han Chinese women.
Collapse
Affiliation(s)
- Ji-peng Wan
- Department of Obstetrics and Gynecology, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Han Zhao
- Center for Reproductive Medicine, Provincial Hospital Affiliated to Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key Laboratory for Reproductive Endocrinology of Ministry of Education, Jinan, China
- Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China
| | - Tao Li
- Center for Reproductive Medicine, Provincial Hospital Affiliated to Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key Laboratory for Reproductive Endocrinology of Ministry of Education, Jinan, China
- Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China
| | - Chang-zhong Li
- Department of Obstetrics and Gynecology, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Xie-tong Wang
- Department of Obstetrics and Gynecology, Provincial Hospital Affiliated to Shandong University, Jinan, China
- * E-mail:
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Provincial Hospital Affiliated to Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key Laboratory for Reproductive Endocrinology of Ministry of Education, Jinan, China
- Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China
- Center for Reproductive Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
45
|
Villa PM, Hämäläinen E, Mäki A, Räikkönen K, Pesonen AK, Taipale P, Kajantie E, Laivuori H. Vasoactive agents for the prediction of early- and late-onset preeclampsia in a high-risk cohort. BMC Pregnancy Childbirth 2013; 13:110. [PMID: 23663420 PMCID: PMC3661359 DOI: 10.1186/1471-2393-13-110] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 04/29/2013] [Indexed: 11/24/2022] Open
Abstract
Background To evaluate the soluble fms-like tyrosine kinase-1 (sFlt-1), placental growth factor (PlGF), and sFlt-1/PlGF ratio for the prediction of early- and late-onset preeclampsia in a high-risk cohort. Methods We studied serial serum samples collected prospectively at 12 + 0 - 14 + 0, 18 + 0 - 20 + 0, and 26 + 0 - 28 + 0 weeks + days of gestation in 6 women who developed early-onset preeclampsia (before 34 weeks of gestation) and in 21 women who developed late-onset preeclampsia (after 34 weeks of gestation) with automated ElecSys 2010 immunoanalyzer (Roche Diagnostics, Germany). Twenty-six high-risk women and 53 women without risk factors with normal pregnancies served as controls. Results Serum PlGF concentrations were lower at 18 + 0 to 20 + 0, and 26 + 0 to 28 + 0 weeks of gestation in women who developed early-onset preeclampsia compared to women who developed late-onset preeclampsia and to controls (p < 0.05 for all comparisons). At 18 + 0 to 20 + 0 weeks of gestation area under the receiver-operating characteristic curve (AUC) for serum PlGF was 99.8% (p = 0.0007, 95% CI 99.0-100.0). At 26 + 0 to 28 + 0 weeks of gestation serum sFlt-1/PlGF ratio explicitly detects those women who developed early-onset preeclampsia (AUC 100.0%, p = 0.0007, 95% CI 100–100). Amongst women with late-onset preeclampsia, those who developed severe form of the disease (N = 8) had significantly higher serum sFlt-1 concentrations at all three timepoints (p = 0.004, p = 0.006, and p = 0.003, respectively) compared to women with non-severe form (N = 13). Conclusions Low serum PlGF concentration predicts early-onset preeclampsia from the second trimester and elevated serum sFlt-1/PlGF ratio from 26 to 28 weeks of gestation. Elevated serum sFlt-1 concentration in the first trimester in women who later develop late-onset, severe preeclampsia may suggest different etiology compared to the late-onset non-severe form of the disease.
Collapse
Affiliation(s)
- Pia M Villa
- The Research Programs Unit, Women's Health, University of Helsinki, and Department of Obstetrics and Gynaecology, Helsinki University Central Hospital, Helsinki, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Yang J, Shang J, Zhang S, Li H, Liu H. The role of the renin-angiotensin-aldosterone system in preeclampsia: genetic polymorphisms and microRNA. J Mol Endocrinol 2013; 50:R53-66. [PMID: 23369849 DOI: 10.1530/jme-12-0216] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The compensatory alterations in the rennin-angiotensin-aldosterone system (RAAS) contribute to the salt-water balance and sufficient placental perfusion for the subsequent well-being of the mother and fetus during normal pregnancy and is characterized by an increase in almost all the components of RAAS. Preeclampsia, however, breaks homeostasis and leads to a disturbance of this delicate equilibrium in RAAS both for circulation and the uteroplacental unit. Despite being a major cause for maternal and neonatal morbidity and mortality, the pathogenesis of preeclampsia remains elusive, where RAAS has been long considered to be involved. Epidemiological studies have indicated that preeclampsia is a multifactorial disease with a strong familial predisposition regardless of variations in ethnic, socioeconomic, and geographic features. The heritable allelic variations, especially the genetic polymorphisms in RAAS, could be the foundation for the genetics of preeclampsia and hence are related to the development of preeclampsia. Furthermore, at a posttranscriptional level, miRNA can interact with the targeted site within the 3'-UTR of the RAAS gene and thereby might participate in the regulation of RAAS and the pathology of preeclampsia. In this review, we discuss the recent achievements of genetic polymorphisms, as well as the interactions between maternal and fetal genotypes, and miRNA posttranscriptional regulation associated with RAAS in preeclampsia. The results are controversial but utterly inspiring and attractive in terms of potential prognostic significance. Although many studies suggest positive associations with genetic mutations and increased risk for preeclampsia, more meticulously designed large-scale investigations are needed to avoid the interference from different variations.
Collapse
Affiliation(s)
- Jie Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, People's Republic of China
| | | | | | | | | |
Collapse
|
47
|
Abstract
The human genome contains a hidden and large layer of biologic information that is not accessible by proteomic or metabolic methods. Insight into the nature, size, function and importance of this information is increasing rapidly. This additional layer of information includes non-coding RNA and DNA and can be retrieved and analyzed using nucleic acids that circulate in the maternal plasma during pregnancy, originate from the developing placenta and provide information on fetal well being. This review explains why, when and how fetal information as carried on and provided by the placental DNA and RNA molecules circulating in the plasma of pregnant women can be explored to understand and to analyze the primary placental processes, that underlie pre-eclampsia and related disorders.
Collapse
Affiliation(s)
- Cees Bm Oudejans
- VU University Medical Center, Department of Clinical Chemistry, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands +31 20 444 3867 ; +31 20 444 3895 ;
| |
Collapse
|
48
|
A follow-up linkage study of Finnish pre-eclampsia families identifies a new fetal susceptibility locus on chromosome 18. Eur J Hum Genet 2013; 21:1024-6. [PMID: 23386034 DOI: 10.1038/ejhg.2013.6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 12/20/2012] [Accepted: 01/10/2013] [Indexed: 11/08/2022] Open
Abstract
Pre-eclampsia is a common vascular disorder of pregnancy. It originates in the placenta and targets the maternal endothelium. According to epidemiological research, >50% of the liability to this disorder can be accounted for by genetic factors. Both maternal and fetal genes contribute to the risk, but especially the fetal genetic risk profile is still poorly understood. We have previously detected linkage signals in multiplex Finnish families on chromosomes 2p25, 4q32, and 9p13 using maternal phenotypes. We performed a linkage analysis using updated maternal phenotypes and an unprecedented linkage analysis using fetal phenotypes. Markers genotyped were available from 237 individuals in 15 Finnish families, including 72 affected mothers and 49 affected fetuses. The MERLIN software was used for sample and marker quality control and linkage analysis. The results were compared against the original ones obtained by using the GENEHUNTER 2.1 software. The previous identification of the maternal susceptibility locus to a genetic location at 21.70 cM near marker D2S168 on chromosome 2 was confirmed by using both maternal and fetal phenotypes (maternal non-parametric linkage (NPL) score 3.79, P=0.00008, LOD (logarithm (base 10) of odds)=2.20 and fetal NPL score 2.95, P=0.002, LOD=1.71). As a novel finding, we present a suggestive linkage to chromosome 18 at 86.80 cM near marker D18S64 (NPL score 2.51, P=0.006, LOD=1.20) using the fetal phenotype. We propose that chromosome 18 may harbor a new fetal susceptibility locus for pre-eclampsia.
Collapse
|
49
|
Elevated expression of KiSS-1 in placenta of Chinese women with early-onset preeclampsia. PLoS One 2012; 7:e48937. [PMID: 23145030 PMCID: PMC3493594 DOI: 10.1371/journal.pone.0048937] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 10/01/2012] [Indexed: 11/25/2022] Open
Abstract
Preeclampsia (PE) is a heterogeneous syndrome affecting 2% to 8% of all pregnancies and is the world’s leading cause of fetal and maternal morbidity and mortality. In many cases of PE, shallow trophoblast invasion results in inappropriate maternal spiral artery remodeling and impaired placental function. Multiple genes have been implicated in trophoblast invasion, among which are KiSS-1 and GPR54. The gene product of KiSS-1 is metastin, which is a ligand for the receptor GPR54. Both metastin and GPR54 are expressed in the placenta of normal pregnancy and have been implicated in modulating trophoblast invasion through inhibiting migration of trophoblast cells. We have previously reported that the expression level of KiSS-1 was higher in trophoblasts from women with preeclampsia as compared to normal controls. Here, using quantitative RT-PCR, Western blot analysis and immunohistochemistry, we extend our analysis to demonstrate that elevated KiSS-1 expression occurs only in early-onset preeclampsia (ePE) and not late-onset preeclampsia (lPE). However, no difference in the expression levels of GPR54 is observed between ePE, lPE, and normal controls. Further, we show that KiSS-1 expression is also increased in placenta of intrauterine death and birth asphyxia in comparison to normal newborns of ePE and lPE. Our findings suggest that aberrant upregulation of KiSS-1 expression may contribute to the underlying mechanism of ePE as well as birth asphyxia.
Collapse
|
50
|
Villa PM, Kajantie E, Räikkönen K, Pesonen AK, Hämäläinen E, Vainio M, Taipale P, Laivuori H. Aspirin in the prevention of pre-eclampsia in high-risk women: a randomised placebo-controlled PREDO Trial and a meta-analysis of randomised trials. BJOG 2012; 120:64-74. [DOI: 10.1111/j.1471-0528.2012.03493.x] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|