51
|
Human Genetic Adaptation to High Altitude: Evidence from the Andes. Genes (Basel) 2019; 10:genes10020150. [PMID: 30781443 PMCID: PMC6410003 DOI: 10.3390/genes10020150] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/29/2019] [Accepted: 02/11/2019] [Indexed: 12/31/2022] Open
Abstract
Whether Andean populations are genetically adapted to high altitudes has long been of interest. Initial studies focused on physiological changes in the O₂ transport system that occur with acclimatization in newcomers and their comparison with those of long-resident Andeans. These as well as more recent studies indicate that Andeans have somewhat larger lung volumes, narrower alveolar to arterial O₂ gradients, slightly less hypoxic pulmonary vasoconstrictor response, greater uterine artery blood flow during pregnancy, and increased cardiac O2 utilization, which overall suggests greater efficiency of O₂ transfer and utilization. More recent single nucleotide polymorphism and whole-genome sequencing studies indicate that multiple gene regions have undergone recent positive selection in Andeans. These include genes involved in the regulation of vascular control, metabolic hemostasis, and erythropoiesis. However, fundamental questions remain regarding the functional links between these adaptive genomic signals and the unique physiological attributes of highland Andeans. Well-designed physiological and genome association studies are needed to address such questions. It will be especially important to incorporate the role of epigenetic processes (i.e.; non-sequence-based features of the genome) that are vital for transcriptional responses to hypoxia and are potentially heritable across generations. In short, further exploration of the interaction among genetic, epigenetic, and environmental factors in shaping patterns of adaptation to high altitude promises to improve the understanding of the mechanisms underlying human adaptive potential and clarify its implications for human health.
Collapse
|
52
|
Komiya H, Miyoshi C, Iwasaki K, Hotta-Hirashima N, Ikkyu A, Kanno S, Honda T, Gosho M, Hamada H, Satoh T, Fukamizu A, Funato H, Yanagisawa M. Sleep/Wake Behaviors in Mice During Pregnancy and Pregnancy-Associated Hypertensive Mice. Sleep 2019; 41:4773862. [PMID: 29309677 DOI: 10.1093/sleep/zsx209] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 12/06/2017] [Indexed: 11/13/2022] Open
Abstract
Study Objectives In humans and other mammals, sleep is altered during pregnancy. However, no studies have been conducted on sleep/wakefulness during pregnancy in mice. In this study, we examined sleep/wakefulness in female C57BL/6 mice during pregnancy. We also examined sleep/wake behaviors in an animal model of preeclampsia, pregnancy-associated hypertensive (PAH) mice, in which increased angiotensin causes hypertension. Methods Sleep/wake behaviors of female C57BL/6 and PAH mice were examined based on electroencephalogram (EEG) or electromyogram recordings before, during, and after pregnancy. To examine whether high blood pressure disrupts the integrity of the blood-brain barrier in PAH mice, Evans blue dye was injected intravenously. Angiotensin II receptor blocker (olmesartan)-administered PAH mice and female Tsukuba hypertensive mice were also examined. Results C57BL/6 mice showed a decreased total wake time and increased nonrapid eye movement (NREM) sleep time during late pregnancy. Rapid eye movement (REM) sleep time did not change during the course of pregnancy. PAH mice exhibited a general slowing of EEG during late pregnancy and subsequently returned to apparently normal sleep/wakefulness after delivery. All PAH mice exhibited multiple focal leakages of Evans blue dye in the brain. Spike-and-wave discharges were observed in 50% of PAH mice. Olmesartan-administered PAH mice did not show general slowing of EEG. Tsukuba hypertensive mice showed a normal time spent in wakefulness and NREM sleep and a decreased total REM sleep time. Conclusions This study showed pregnant-stage-specific changes in sleep/wakefulness in C57BL/6 mice. Furthermore, PAH mice may be useful as an animal model for eclampsia.
Collapse
Affiliation(s)
- Haruna Komiya
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Chika Miyoshi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kanako Iwasaki
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Noriko Hotta-Hirashima
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Aya Ikkyu
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Satomi Kanno
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takato Honda
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Masahiko Gosho
- Department of Biostatistics, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiromi Hamada
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Toyomi Satoh
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Akiyoshi Fukamizu
- Life Science Center, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiromasa Funato
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Anatomy, Faculty of Medicine, Toho University, Ota-ku, Tokyo, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan.,Life Science Center, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Anatomy, Faculty of Medicine, Toho University, Ota-ku, Tokyo, Japan.,Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
53
|
Jung YW, Shim JI, Shim SH, Shin YJ, Shim SH, Chang SW, Cha DH. Global gene expression analysis of cell-free RNA in amniotic fluid from women destined to develop preeclampsia. Medicine (Baltimore) 2019; 98:e13971. [PMID: 30653101 PMCID: PMC6370049 DOI: 10.1097/md.0000000000013971] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Preeclampsia (PE) is a disorder specific to pregnancy characterized by new-onset hypertension and proteinuria after 20 weeks of gestation. There is no definite treatment for PE except delivery of the placenta. The purpose of this study was to elucidate the biological pathways involved in the development of PE and to discover a novel biomarker for PE by performing global gene expression analysis of amniotic fluid cell-free RNA.The participants were recruited from the Department of Obstetrics and Gynecology of CHA Gangnam Medical Center (Seoul, Korea) between March 2014 and February 2015. Eight samples were collected from 8 subjects at second trimester who were later diagnosed with PE. From the amniotic fluid samples, cell-free RNA extraction was performed and gene expression was analyzed using the GeneChip PrimeView Array. Transcriptome data previously analyzed by our group from 9 euploid mid-trimester amniotic fluid samples were used as the control for comparative analysis. Functional analysis of the probe sets was performed using the online Database for Annotation, Visualization, and Integrated Discovery (DAVID) toolkit 6.7.We identified 1841 differentially expressed genes (DEGs) between the PE group and the control. Of these, 1557 genes were upregulated in the PE group, while 284 genes were upregulated in the control. The functional annotation of DEGs identified specific enriched functions such as "transport," "signal transduction," and "stress response." Functional annotation clustering with enriched genes in the PE group revealed that translation-related genes, cell-cell adhesion genes, and immune-related genes were enriched. KEGG pathway analysis showed that several biological pathways, including the ribosome pathway and various immune pathways, were dysregulated. Several genes, including RPS29, IGF-2, and UBC, were significantly upregulated in PE, up to tenfold.This study provides the first genome-wide expression analysis of amniotic fluid cell-free RNA in PE. The results showed that gene expression involving the ribosome pathway and immunologic pathways are dysregulated in PE. Our results will aid in understanding the underlying pathogenesis of PE.
Collapse
Affiliation(s)
- Yong Wook Jung
- Department of Obstetrics and Gynecology, CHA Gangnam Medical Center
| | - Jung In Shim
- Department of Obstetrics and Gynecology, CHA Gangnam Medical Center
| | - So Hyun Shim
- Department of Obstetrics and Gynecology, CHA Gangnam Medical Center
| | - Yun-jeong Shin
- Genetics Laboratory, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul
| | - Sung Han Shim
- Genetics Laboratory, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul
| | - Sung Woon Chang
- Department of Obstetrics and Gynecology, CHA Bundang Medical Center, CHA University, Seongnam-si, Republic of Korea
| | - Dong Hyun Cha
- Department of Obstetrics and Gynecology, CHA Gangnam Medical Center
- Genetics Laboratory, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul
| |
Collapse
|
54
|
Maher GM, McCarthy FP, McCarthy CM, Kenny LC, Kearney PM, Khashan AS, O'Keeffe GW. A perspective on pre-eclampsia and neurodevelopmental outcomes in the offspring: Does maternal inflammation play a role? Int J Dev Neurosci 2018; 77:69-76. [PMID: 30391740 DOI: 10.1016/j.ijdevneu.2018.10.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/31/2018] [Accepted: 10/31/2018] [Indexed: 10/28/2022] Open
Abstract
Pre-eclampsia is a leading cause of maternal death and maternal and perinatal morbidity. Whilst the clinical manifestations of pre-eclampsia often occur in late pregnancy, the molecular events leading into the onset of this disease are thought to originate in early pregnancy and result in insufficient placentation. Although the causative molecular basis of pre-eclampsia remains poorly understood, maternal inflammation is recognised as a core clinical feature. While the adverse effects of pre-eclampsia on maternal and fetal health in pregnancy is well-recognised, the long-term impact of pre-eclampsia exposure on the risk of autism spectrum disorder (ASD) in exposed offspring is a topic of on-going debate. In particular, a recent systematic review has reported an association between exposure to pre-eclampsia and increased risk of ASD, however the molecular basis of this association is unknown. Here we review recent evidence for; 1) maternal inflammation in pre-eclampsia; 2) epidemiological evidence for alterations in neurodevelopmental outcomes in offspring exposed to pre-eclampsia; 3) long-term changes in the brains of offspring exposed to pre-eclampsia; and 4) how maternal inflammation may lead to altered neurodevelopmental outcomes in pre-eclampsia exposed offspring. Finally, we discuss the implications of this for the development of future studies in this field.
Collapse
Affiliation(s)
- Gillian M Maher
- School of Public Health, Western Gateway Building, University College Cork, Cork, Ireland.,The Irish Centre for Fetal and Neonatal Translational Research (INFANT), Cork University Maternity Hospital and University College Cork, Cork, Ireland
| | - Fergus P McCarthy
- The Irish Centre for Fetal and Neonatal Translational Research (INFANT), Cork University Maternity Hospital and University College Cork, Cork, Ireland.,Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, University College Cork, Ireland
| | - Cathal M McCarthy
- The Irish Centre for Fetal and Neonatal Translational Research (INFANT), Cork University Maternity Hospital and University College Cork, Cork, Ireland.,Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland
| | - Louise C Kenny
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, United Kingdom
| | - Patricia M Kearney
- School of Public Health, Western Gateway Building, University College Cork, Cork, Ireland
| | - Ali S Khashan
- School of Public Health, Western Gateway Building, University College Cork, Cork, Ireland.,The Irish Centre for Fetal and Neonatal Translational Research (INFANT), Cork University Maternity Hospital and University College Cork, Cork, Ireland
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience and Cork Neuroscience Centre, Western Gateway Building, University College Cork, Cork, Ireland
| |
Collapse
|
55
|
Kose S, Tuna G, Nuriyeva G, Altunyurt S, Islekel GH, Doğan OE. A prospective cohort study on the prediction of the diagnosis-to-delivery time in preeclamptic pregnancies: should the sFlt-1/PlGF ratio be added to routine evaluations? Arch Gynecol Obstet 2018; 298:911-920. [DOI: 10.1007/s00404-018-4903-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 09/06/2018] [Indexed: 11/24/2022]
|
56
|
Than NG, Romero R, Tarca AL, Kekesi KA, Xu Y, Xu Z, Juhasz K, Bhatti G, Leavitt RJ, Gelencser Z, Palhalmi J, Chung TH, Gyorffy BA, Orosz L, Demeter A, Szecsi A, Hunyadi-Gulyas E, Darula Z, Simor A, Eder K, Szabo S, Topping V, El-Azzamy H, LaJeunesse C, Balogh A, Szalai G, Land S, Torok O, Dong Z, Kovalszky I, Falus A, Meiri H, Draghici S, Hassan SS, Chaiworapongsa T, Krispin M, Knöfler M, Erez O, Burton GJ, Kim CJ, Juhasz G, Papp Z. Integrated Systems Biology Approach Identifies Novel Maternal and Placental Pathways of Preeclampsia. Front Immunol 2018; 9:1661. [PMID: 30135684 PMCID: PMC6092567 DOI: 10.3389/fimmu.2018.01661] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/04/2018] [Indexed: 12/13/2022] Open
Abstract
Preeclampsia is a disease of the mother, fetus, and placenta, and the gaps in our understanding of the complex interactions among their respective disease pathways preclude successful treatment and prevention. The placenta has a key role in the pathogenesis of the terminal pathway characterized by exaggerated maternal systemic inflammation, generalized endothelial damage, hypertension, and proteinuria. This sine qua non of preeclampsia may be triggered by distinct underlying mechanisms that occur at early stages of pregnancy and induce different phenotypes. To gain insights into these molecular pathways, we employed a systems biology approach and integrated different "omics," clinical, placental, and functional data from patients with distinct phenotypes of preeclampsia. First trimester maternal blood proteomics uncovered an altered abundance of proteins of the renin-angiotensin and immune systems, complement, and coagulation cascades in patients with term or preterm preeclampsia. Moreover, first trimester maternal blood from preterm preeclamptic patients in vitro dysregulated trophoblastic gene expression. Placental transcriptomics of women with preterm preeclampsia identified distinct gene modules associated with maternal or fetal disease. Placental "virtual" liquid biopsy showed that the dysregulation of these disease gene modules originates during the first trimester. In vitro experiments on hub transcription factors of these gene modules demonstrated that DNA hypermethylation in the regulatory region of ZNF554 leads to gene down-regulation and impaired trophoblast invasion, while BCL6 and ARNT2 up-regulation sensitizes the trophoblast to ischemia, hallmarks of preterm preeclampsia. In summary, our data suggest that there are distinct maternal and placental disease pathways, and their interaction influences the clinical presentation of preeclampsia. The activation of maternal disease pathways can be detected in all phenotypes of preeclampsia earlier and upstream of placental dysfunction, not only downstream as described before, and distinct placental disease pathways are superimposed on these maternal pathways. This is a paradigm shift, which, in agreement with epidemiological studies, warrants for the central pathologic role of preexisting maternal diseases or perturbed maternal-fetal-placental immune interactions in preeclampsia. The description of these novel pathways in the "molecular phase" of preeclampsia and the identification of their hub molecules may enable timely molecular characterization of patients with distinct preeclampsia phenotypes.
Collapse
Affiliation(s)
- Nandor Gabor Than
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, United States
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, United States
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
| | - Adi Laurentiu Tarca
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Computer Science, College of Engineering, Wayne State University, Detroit, MI, United States
| | - Katalin Adrienna Kekesi
- Laboratory of Proteomics, Department of Physiology and Neurobiology, ELTE Eotvos Lorand University, Budapest, Hungary
| | - Yi Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
| | - Zhonghui Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard University, Boston, MA, United States
| | - Kata Juhasz
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gaurav Bhatti
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
| | | | - Zsolt Gelencser
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Janos Palhalmi
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | | | - Balazs Andras Gyorffy
- Laboratory of Proteomics, Department of Physiology and Neurobiology, ELTE Eotvos Lorand University, Budapest, Hungary
| | - Laszlo Orosz
- Department of Obstetrics and Gynaecology, University of Debrecen, Debrecen, Hungary
| | - Amanda Demeter
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Anett Szecsi
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Eva Hunyadi-Gulyas
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Zsuzsanna Darula
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Attila Simor
- Laboratory of Proteomics, Department of Physiology and Neurobiology, ELTE Eotvos Lorand University, Budapest, Hungary
| | - Katalin Eder
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Szilvia Szabo
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
- Department of Morphology and Physiology, Semmelweis University, Budapest, Hungary
| | - Vanessa Topping
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
| | - Haidy El-Azzamy
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
| | - Christopher LaJeunesse
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
| | - Andrea Balogh
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gabor Szalai
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Susan Land
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
| | - Olga Torok
- Department of Obstetrics and Gynaecology, University of Debrecen, Debrecen, Hungary
| | - Zhong Dong
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
| | - Ilona Kovalszky
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Andras Falus
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | | | - Sorin Draghici
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
- Department of Clinical and Translational Science, Wayne State University, Detroit, MI, United States
| | - Sonia S. Hassan
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | | | - Martin Knöfler
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Offer Erez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Soroka University Medical Center School of Medicine, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Graham J. Burton
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Chong Jai Kim
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Pathology, Asan Medical Center, University of Ulsan, Seoul, South Korea
| | - Gabor Juhasz
- Laboratory of Proteomics, Department of Physiology and Neurobiology, ELTE Eotvos Lorand University, Budapest, Hungary
| | - Zoltan Papp
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
| |
Collapse
|
57
|
Combination of Genetic Markers and Age Effectively Facilitates the Identification of People with High Risk of Preeclampsia in the Han Chinese Population. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4808046. [PMID: 30112393 PMCID: PMC6077688 DOI: 10.1155/2018/4808046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/15/2018] [Accepted: 06/11/2018] [Indexed: 01/03/2023]
Abstract
Objective This study aimed to analyze the possible association between known genetic risks and preeclampsia in a Han Chinese population. Methods A total of 156 patients with preeclampsia and 286 healthy Han Chinese women were enrolled and genotyped for 27 genetic alleles associated with preeclampsia in different populations. The association between the genotypes of the individual alleles and preeclampsia and the possible interaction among the alleles were analyzed. Finally logistic models were trained with the genotypes of possible alleles contributing to preeclampsia. Results Seven alleles were significantly or marginally significantly associated with preeclampsia, which involved six genes (rs4762 in AGT, rs1800896 in IL-10, rs1800629 and rs1799724 in TNFα, rs2070744 in NOS3, rs7412 in APOE, and rs2549782 in ERAP2). A multilocus interaction analysis further disclosed an interaction among seven alleles. A logistic model showing individual or synergetic contribution to preeclampsia could reach ~0.67 preeclampsia prediction accuracy in the Han Chinese population, while integration of age information could improve the performance to ~0.75 accuracy using a fivefold training-testing evaluation strategy. Conclusions The genetic factors were closely associated with preeclampsia in the Han Chinese population despite large ethnicity heterogeneity. The genotypes of different alleles also had synergetic interactions.
Collapse
|
58
|
Kuo CY, Guo T, Cabrera-Luque J, Arumugasaamy N, Bracaglia L, Garcia-Vivas A, Santoro M, Baker H, Fisher J, Kim P. Placental basement membrane proteins are required for effective cytotrophoblast invasion in a three-dimensional bioprinted placenta model. J Biomed Mater Res A 2018; 106:1476-1487. [PMID: 29368378 PMCID: PMC5924478 DOI: 10.1002/jbm.a.36350] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/11/2018] [Accepted: 01/19/2018] [Indexed: 11/06/2022]
Abstract
Fetal cytotrophoblast invasion of maternal decidual vasculature is necessary to normal pregnancy. In preeclampsia, there is shallow invasion and abnormal remodeling of the uterine vasculature that lead to significant maternal and perinatal morbidity and mortality. The placental basement membrane (BM) proteins (e.g., laminin and collagen) has been implicated in the development of placenta while the level of laminin is significantly lower in preeclampsia. However, there are very limited studies, if any, on the effect of extracellular matrix (ECM) microenvironment on the invasion of cytotrophoblast. In this study, we hypothesized that placental BM proteins are required for effective cytotrophoblast invasion. Using proteomics, we found that more than 80% of ECM proteins in placental basal plate (pECM) were BM proteins. In addition to upregulating expressions of MMP2 (1.5-fold) and MMP9 (6.3-fold), pECM significantly increased the motility rates of cytotrophoblasts by 13-fold (from 5.60 ± 0.95 to 75.5 ± 21.8 µm/day) to achieve an effective invasion rate that was comparable to in vivo results. Treatments with PI3K inhibitors completely removed the pECM-enhanced invasive phenotypes and genotypes of cytotrophoblasts, suggesting its dominant role in cytotrophoblast-ECM interactions. Our results described, for the first time, the substantial effects of the ECM microenvironment on regulating cytotrophoblast invasion, an area that is less investigated but appear to be critical in the pathogenesis of preeclampsia. Moreover, the approach presented in this work that fabricates organ models with organ-specific ECM can be an attractive option to screen and develop novel therapeutics and biomarkers not only in preeclampsia but also other diseases such as cancer metastasis. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 1476-1487, 2018.
Collapse
Affiliation(s)
- Che-Ying Kuo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Ting Guo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Juan Cabrera-Luque
- Center for Genetic Medicine, Children’s National Health System, Washington, DC
| | - Navein Arumugasaamy
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Laura Bracaglia
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Amy Garcia-Vivas
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Marco Santoro
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Hannah Baker
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - John Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Peter Kim
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- School of Medicine and Health Sciences, The George Washington University, Washington, DC
| |
Collapse
|
59
|
Li X, Wu C, Shen Y, Wang K, Tang L, Zhou M, Yang M, Pan T, Liu X, Xu W. Ten-eleven translocation 2 demethylates the MMP9 promoter, and its down-regulation in preeclampsia impairs trophoblast migration and invasion. J Biol Chem 2018; 293:10059-10070. [PMID: 29773648 DOI: 10.1074/jbc.ra117.001265] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/22/2018] [Indexed: 12/24/2022] Open
Abstract
Preeclampsia is the most common clinical disorder in pregnancy and might result from disordered uterine environments caused by epigenetic modifications, including deregulation of DNA methylation/demethylation. Recent research has indicated that 5-hydroxymethylcytosine (5hmC), a DNA base derived from 5-methylcytosine (5mC) via oxidation by ten-eleven translocation (TET) enzymes, is involved in DNA methylation-related plasticity. Here, we report that TET2 expression and 5hmC abundance are significantly altered in the placentas from preeclampsia patients. shRNA-mediated TET2 knockdown (shTET2) reduced trophoblast migration and invasion when cultured in Matrigel. Both real-time PCR of matrix metalloproteinase (MMP)-related transcripts and a human angiogenesis antibody array indicated that TET2 knockdown in trophoblasts inhibits the expression of MMP transcript, of which MMP9 represented one of the most significant TET2 downstream targets. Using an established shTET2 HTR-8/SVneo cell model, we further confirmed alterations of 5hmC levels and MMP9 expression at both mRNA and protein levels. In particular, we found that TET2 bound to and removed 5mC modifications at the MMP9 promoter region. Interestingly, in TET2 knockdown cells, both MMP9 expression and the compromised trophoblast phenotype could be rescued by vitamin C, an activator of TET enzyme activity. Finally, TET2 expression correlated with MMP9 levels in placenta samples from the preeclampsia patients, indicating that TET2 deregulation is critically involved in the pathogenesis of preeclampsia through down-regulation of MMP9 expression. Our findings highlight a critical role of TET2 in regulating trophoblast cell migration through demethylation at the MMP9 promoter, and suggest that down-regulation of the TET2-MMP9-mediated pathway contributes to preeclampsia pathogenesis.
Collapse
Affiliation(s)
- Xiaoliang Li
- From the Joint Laboratory of Reproductive Medicine, SCU-CUHK, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Department of Obstetric and Gynecologic Diseases, West China Second University Hospital, Sichuan University, Chengdu 610041, China, and.,Key Laboratory of Southwest China Wildlife Resource Conservation (China West Normal University), Ministry of Education, Nanchong 637009 China
| | - Chunlian Wu
- Key Laboratory of Southwest China Wildlife Resource Conservation (China West Normal University), Ministry of Education, Nanchong 637009 China
| | - Ying Shen
- From the Joint Laboratory of Reproductive Medicine, SCU-CUHK, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Department of Obstetric and Gynecologic Diseases, West China Second University Hospital, Sichuan University, Chengdu 610041, China, and
| | - Ke Wang
- From the Joint Laboratory of Reproductive Medicine, SCU-CUHK, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Department of Obstetric and Gynecologic Diseases, West China Second University Hospital, Sichuan University, Chengdu 610041, China, and
| | - Li Tang
- From the Joint Laboratory of Reproductive Medicine, SCU-CUHK, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Department of Obstetric and Gynecologic Diseases, West China Second University Hospital, Sichuan University, Chengdu 610041, China, and
| | - Mi Zhou
- From the Joint Laboratory of Reproductive Medicine, SCU-CUHK, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Department of Obstetric and Gynecologic Diseases, West China Second University Hospital, Sichuan University, Chengdu 610041, China, and
| | - Ming Yang
- From the Joint Laboratory of Reproductive Medicine, SCU-CUHK, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Department of Obstetric and Gynecologic Diseases, West China Second University Hospital, Sichuan University, Chengdu 610041, China, and
| | - Tianying Pan
- From the Joint Laboratory of Reproductive Medicine, SCU-CUHK, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Department of Obstetric and Gynecologic Diseases, West China Second University Hospital, Sichuan University, Chengdu 610041, China, and
| | - Xinghui Liu
- Department of Obstetric and Gynecologic Diseases, West China Second University Hospital, Sichuan University, Chengdu 610041, China, and
| | - Wenming Xu
- From the Joint Laboratory of Reproductive Medicine, SCU-CUHK, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China, .,Department of Obstetric and Gynecologic Diseases, West China Second University Hospital, Sichuan University, Chengdu 610041, China, and
| |
Collapse
|
60
|
Nair TM. Statistical and artificial neural network-based analysis to understand complexity and heterogeneity in preeclampsia. Comput Biol Chem 2018; 75:222-230. [PMID: 29859381 DOI: 10.1016/j.compbiolchem.2018.05.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 04/11/2018] [Accepted: 05/09/2018] [Indexed: 12/11/2022]
Abstract
Preeclampsia is a pregnancy associated disease. It is characterized by high blood pressure and symptoms that are indicative of damage to other organ systems, most often involving the liver and kidneys. If left untreated, the condition could be fatal to mother and baby. This makes it important to delineate the complexities associated with the disease at a molecular level that would help develop methods for early diagnosis. In microarray-based studies, Textoris et al. and Mirzakhani et al. have analyzed the transcriptome with a view to identify biomarkers for preeclampsia. The current study has extensively analyzed these microarray data sets to understand the complexity and heterogeneity associated with preeclampsia. A statistical multiple comparisons-based approach has been used to identify features capable of distinguishing preeclampsia from normotensive cases. These features were then used to build an artificial neural network-based machine learning model that successfully classified the samples. Further, the machine learning model was used to delineate features critical for its internal representation by extending the calliper randomization approach to the analysis of microarray data. Functional analysis of the features identified by the calliper randomization approach revealed pathways that could be crucially involved in the mechanism of the underlying disease. Biological processes associated with the features identified have revealed among others, genes involved in reproductive processes to be differentially expressed.
Collapse
Affiliation(s)
- T Murlidharan Nair
- Department of Biological Sciences, Indiana University South Bend, South Bend, IN, United States; Department of Computer Science/Informatics, Indiana University South Bend, South Bend, IN, United States.
| |
Collapse
|
61
|
Leijnse JEW, de Heus R, de Jager W, Rodenburg W, Peeters LLH, Franx A, Eijkelkamp N. First trimester placental vascularization and angiogenetic factors are associated with adverse pregnancy outcome. Pregnancy Hypertens 2018; 13:87-94. [PMID: 30177079 DOI: 10.1016/j.preghy.2018.04.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 02/11/2018] [Accepted: 04/11/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Hypertensive disorders, fetal growth restriction and preterm birth are major obstetrical complications and are related to impaired placentation. Early identification of impaired placentation can advance clinical care by preventing or postpone adverse pregnancy outcome. OBJECTIVES Determine whether sonographic assessed placental vascular development and concomitant changes in inflammation- and/or angiogenesis-related serumproteins differ in the first trimester between uncomplicated pregnancies and pregnancies with adverse outcome. STUDY DESIGN This prospective longitudinal study defines adverse pregnancy outcome as conditions associated with impaired placentation; fetal growth restriction, hypertensive disorder, preterm birth and placental abruption. The vascularization index, flow index, vascularization flow index and placental volume were determined at 8, 10 and 12 weeks pregnancy from 64 women using 3D power Doppler. Serum levels were analyzed for Angiopoetin-1 and -2, Leptin, VEGF-R, VEGF, and EGF. RESULTS The vascularization index and vascular flow index increased in uneventful pregnancies with almost 50% between 8 and 12 weeks, resulting in a ∼50% higher vascularization index at 12 weeks compared to women with an adverse pregnancy outcome. Women with an adverse pregnancy outcome (n = 13) had significantly lower indices and placental volumes at all time points measured and these indices did not increase between 8 and 12 weeks. Reduced vascular development was associated with increased Angiopoietin-1 levels at 8 and 12 weeks and increased Leptin levels at 8 weeks. CONCLUSIONS Pregnancies with an adverse outcome caused by conditions associated with impaired placentation differ from uneventful pregnancies in having reduced placental vascularization accompanied by elevated circulating levels of Angiopoietin-1 and Leptin already in the first trimester.
Collapse
Affiliation(s)
- Johanna E W Leijnse
- Department of Obstetrics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, 3508 GA Utrecht, The Netherlands.
| | - Roel de Heus
- Department of Obstetrics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, 3508 GA Utrecht, The Netherlands
| | - Wilco de Jager
- Department of Pediatric Immunology, Laboratory of Translational Immunology and Multiplex Core Facility, Wilhelmina Children's Hospital, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands
| | - Wendy Rodenburg
- National Institute for Public Health and Environment (RIVM), 3721 MA Bilthoven, The Netherlands
| | - Louis L H Peeters
- Department of Obstetrics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, 3508 GA Utrecht, The Netherlands
| | - Arie Franx
- Department of Obstetrics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, 3508 GA Utrecht, The Netherlands
| | - Niels Eijkelkamp
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), Wilhelmina Children's Hospital, University Medical Centre Utrecht, 3584 EA Utrecht, The Netherlands
| |
Collapse
|
62
|
Koroglu N, Tola E, Temel Yuksel I, Aslan Cetin B, Turhan U, Topcu G, Dag I. Maternal serum AMP-activated protein kinase levels in mild and severe preeclampsia. J Matern Fetal Neonatal Med 2018; 32:2735-2740. [PMID: 29504441 DOI: 10.1080/14767058.2018.1448774] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJECTIVE To investigate Phosphorylated adenosine monophosphate activated protein kinase (AMPK) levels in healthy pregnant women and pregnant women with preeclampsia (PE). METHODS Twenty-eight women with mild-PE, 22 with severe-PE, and 30 normotensive controls were included in this cross-sectional study. The serum AMPK levels of these patients were analyzed. The patients were followed up to delivery. RESULTS No statistically significant difference was found between the groups for age, gravida, parity, and gestational age at the time the blood samples were obtained (p > .05). No significant difference between the group with mild-PE and the control group was found, while in the severe-PE group, serum AMPK levels were significantly higher relative to both the mild-PE and control groups (p < .001 and p < .001, respectively). No correlation was detected between serum AMPK levels and age, body mass index (BMI), and gestational age at the time the blood samples were collected. A negative correlation was found between AMPK levels and gestational week and birthweight at delivery, while a positive correlation was detected between systolic and diastolic blood pressures and AMPK levels. CONCLUSIONS Serum AMPK was higher in patients with severe-PE compared with healthy pregnant women and patients with PE without severe features so it might be a new biomarker for the prediction of disease and its severity.
Collapse
Affiliation(s)
- Nadiye Koroglu
- a Health Sciences University Kanuni Sultan Suleyman Training and Research Hospital , Istanbul , Turkey
| | - Esra Tola
- b Obstetrics and Gynecology Department , Suleyman Demirel University, Faculty of Medicine , Istanbul , Turkey
| | - Ilkbal Temel Yuksel
- a Health Sciences University Kanuni Sultan Suleyman Training and Research Hospital , Istanbul , Turkey
| | - Berna Aslan Cetin
- a Health Sciences University Kanuni Sultan Suleyman Training and Research Hospital , Istanbul , Turkey
| | - Ugur Turhan
- c Department of Obstetrics and Gynecology , Maternal-Fetal Medicine Unit, Health Sciences University Kanuni Sultan Suleyman Training and Research Hospital, School of Medicine, University of Saglik Bilimleri , Istanbul , Turkey
| | - Goknur Topcu
- a Health Sciences University Kanuni Sultan Suleyman Training and Research Hospital , Istanbul , Turkey
| | - Ismail Dag
- d Eyup State Hospital, Department of Biochemistry , Istanbul , Turkey
| |
Collapse
|
63
|
Angiogenic and Antiangiogenic Markers for Prediction and Risk Classification of Preeclampsia. Clin Obstet Gynecol 2018; 60:134-140. [PMID: 28059847 DOI: 10.1097/grf.0000000000000267] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Preeclampsia is a pregnancy-specific hypertensive disorder with multisystem involvement and is a significant cause of obstetric morbidity and mortality worldwide. A major issue in the treatment of preeclampsia stems from its still significant rates of misclassification and misdiagnosis. Angiogenic factors have been speculated as a possible diagnostic modality due to a perceived imbalance in angiogenesis in preeclampsia. Factors currently studied include soluble fms-like protein kinase 1 and placental growth factor. Because of significant mortality associated with preeclampsia it is felt that both early and accurate diagnosis of preeclampsia is imperative if this disease process is to be treated.
Collapse
|
64
|
Vokalova L, van Breda SV, Ye XL, Huhn EA, Than NG, Hasler P, Lapaire O, Hoesli I, Rossi SW, Hahn S. Excessive Neutrophil Activity in Gestational Diabetes Mellitus: Could It Contribute to the Development of Preeclampsia? Front Endocrinol (Lausanne) 2018; 9:542. [PMID: 30298053 PMCID: PMC6161643 DOI: 10.3389/fendo.2018.00542] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/28/2018] [Indexed: 12/19/2022] Open
Abstract
Gestational diabetes mellitus is a transient form of glucose intolerance occurring during pregnancy. Pregnancies affected by gestational diabetes mellitus are at risk for the development of preeclampsia, a severe life threatening condition, associated with significant feto-maternal morbidity and mortality. It is a risk factor for long-term health in women and their offspring. Pregnancy has been shown to be associated with a subliminal degree of neutrophil activation and tightly regulated generation of neutrophil extracellular traps (NETs). This response is excessive in cases with preeclampsia, leading to the presence of large numbers of NETs in affected placentae. We have recently observed that circulatory neutrophils in cases with gestational diabetes mellitus similarly exhibit an excessive pro-NETotic phenotype, and pronounced placental presence, as detected by expression of neutrophil elastase. Furthermore, exogenous neutrophil elastase liberated by degranulating neutrophils was demonstrated to alter trophoblast physiology and glucose metabolism by interfering with key signal transduction components. In this review we examine whether additional evidence exists suggesting that altered neutrophil activity in gestational diabetes mellitus may contribute to the development of preeclampsia.
Collapse
Affiliation(s)
- Lenka Vokalova
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Institute of Physiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Shane V. van Breda
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Department of Rheumatology, Kantonsspital Aarau, Aarau, Switzerland
| | - Xi Lun Ye
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Evelyn A. Huhn
- Department of Obstetrics, University Women's Hospital Basel, Basel, Switzerland
| | - Nandor G. Than
- Systems Biology of Reproduction Momentum Research Group, Research Centre for Natural Sciences, Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary
| | - Paul Hasler
- Department of Rheumatology, Kantonsspital Aarau, Aarau, Switzerland
| | - Olav Lapaire
- Department of Obstetrics, University Women's Hospital Basel, Basel, Switzerland
| | - Irene Hoesli
- Department of Obstetrics, University Women's Hospital Basel, Basel, Switzerland
| | - Simona W. Rossi
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- *Correspondence: Simona W. Rossi
| | - Sinuhe Hahn
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Sinuhe Hahn
| |
Collapse
|
65
|
Chaiworapongsa T, Romero R, Erez O, Tarca AL, Conde-Agudelo A, Chaemsaithong P, Kim CJ, Kim YM, Kim JS, Yoon BH, Hassan SS, Yeo L, Korzeniewski SJ. The prediction of fetal death with a simple maternal blood test at 20-24 weeks: a role for angiogenic index-1 (PlGF/sVEGFR-1 ratio). Am J Obstet Gynecol 2017; 217:682.e1-682.e13. [PMID: 29037482 PMCID: PMC5951183 DOI: 10.1016/j.ajog.2017.10.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/29/2017] [Accepted: 10/01/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Fetal death is an obstetrical syndrome that annually affects 2.4 to 3 million pregnancies worldwide, including more than 20,000 in the United States each year. Currently, there is no test available to identify patients at risk for this pregnancy complication. OBJECTIVE We sought to determine if maternal plasma concentrations of angiogenic and antiangiogenic factors measured at 24-28 weeks of gestation can predict subsequent fetal death. STUDY DESIGN A case-cohort study was designed to include 1000 randomly selected subjects and all remaining fetal deaths (cases) from a cohort of 4006 women with a singleton pregnancy, enrolled at 6-22 weeks of gestation, in a pregnancy biomarker cohort study. The placentas of all fetal deaths were histologically examined by pathologists who used a standardized protocol and were blinded to patient outcomes. Placental growth factor, soluble endoglin, and soluble vascular endothelial growth factor receptor-1 concentrations were measured by enzyme-linked immunosorbent assays. Quantiles of the analyte concentrations (or concentration ratios) were estimated as a function of gestational age among women who delivered a live neonate but did not develop preeclampsia or deliver a small-for-gestational-age newborn. A positive test was defined as analyte concentrations (or ratios) <2.5th and 10th centiles (placental growth factor, placental growth factor/soluble vascular endothelial growth factor receptor-1 [angiogenic index-1] and placental growth factor/soluble endoglin) or >90th and 97.5th centiles (soluble vascular endothelial growth factor receptor-1 and soluble endoglin). Inverse probability weighting was used to reflect the parent cohort when estimating the relative risk. RESULTS There were 11 fetal deaths and 829 controls with samples available for analysis between 24-28 weeks of gestation. Three fetal deaths occurred <28 weeks and 8 occurred ≥28 weeks of gestation. The rate of placental lesions consistent with maternal vascular underperfusion was 33.3% (1/3) among those who had a fetal death <28 weeks and 87.5% (7/8) of those who had this complication ≥28 weeks of gestation. The maternal plasma angiogenic index-1 value was <10th centile in 63.6% (7/11) of the fetal death group and in 11.1% (92/829) of the controls. The angiogenic index-1 value was <2.5th centile in 54.5% (6/11) of the fetal death group and in 3.7% (31/829) of the controls. An angiogenic index-1 value <2.5th centile had the largest positive likelihood ratio for predicting fetal death >24 weeks (14.6; 95% confidence interval, 7.7-27.7) and a relative risk of 29.1 (95% confidence interval, 8.8-97.1), followed by soluble endoglin >97.5th centile and placental growth factor/soluble endoglin <2.5th, both with a positive likelihood ratio of 13.7 (95% confidence interval, 7.3-25.8) and a relative risk of 27.4 (95% confidence interval, 8.2-91.2). Among women without a fetal death whose plasma angiogenic index-1 concentration ratio was <2.5th centile, 61% (19/31) developed preeclampsia or delivered a small-for-gestational-age neonate; when the 10th centile was used as the cut-off, 37% (34/92) of women had these adverse outcomes. CONCLUSION (1) A maternal plasma angiogenic index-1 value <2.5th centile (0.126) at 24-28 weeks of gestation carries a 29-fold increase in the risk of subsequent fetal death and identifies 55% of subsequent fetal deaths with a false-positive rate of 3.5%; and (2) 61% of women who have a false-positive test result will subsequently experience adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development/National Institutes of Health/Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development/National Institutes of Health/Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI.
| | - Offer Erez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development/National Institutes of Health/Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Obstetrics and Gynecology, Soroka University Medical Center, School of Medicine, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Adi L Tarca
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development/National Institutes of Health/Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Agustin Conde-Agudelo
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development/National Institutes of Health/Department of Health and Human Services, Bethesda, MD, and Detroit, MI
| | - Piya Chaemsaithong
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development/National Institutes of Health/Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Chong Jai Kim
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development/National Institutes of Health/Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yeon Mee Kim
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development/National Institutes of Health/Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Jung-Sun Kim
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development/National Institutes of Health/Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Bo Hyun Yoon
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development/National Institutes of Health/Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sonia S Hassan
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development/National Institutes of Health/Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Lami Yeo
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development/National Institutes of Health/Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Steven J Korzeniewski
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development/National Institutes of Health/Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI
| |
Collapse
|
66
|
Winship A, Dimitriadis E. Interleukin 11 is upregulated in preeclampsia and leads to inflammation and preeclampsia features in mice. J Reprod Immunol 2017; 125:32-38. [PMID: 29195119 DOI: 10.1016/j.jri.2017.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 01/31/2017] [Accepted: 11/24/2017] [Indexed: 12/18/2022]
Abstract
Preeclampsia is a dangerous pregnancy complication, which is often associated with fetal growth restriction and can have serious life-long effects for both mother and baby. While the establishment of the placenta in the first trimester is the sentinel event in the development of preeclampsia little is known of the critical mechanisms of placentation that lead to the syndrome. Locally produced inflammatory cytokines are thought to play a role in the development of preeclampsia. This review summarizes the evidence that interleukin 11 is dysregulated in preeclampsia and contributes to the initiation of preeclampsia via effects on placentation. It discusses the benefits and drawbacks of targeting IL11 as a novel treatment option for preeclampsia.
Collapse
Affiliation(s)
- Amy Winship
- Centre for Reproductive Health, The Hudson Institute of Medical Research, Clayton, 3168, VIC, Australia; Department of Molecular and Translational Medicine, Monash University, Clayton, 3800, VIC, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, 3800, VIC, Australia
| | - Eva Dimitriadis
- Centre for Reproductive Health, The Hudson Institute of Medical Research, Clayton, 3168, VIC, Australia; Department of Molecular and Translational Medicine, Monash University, Clayton, 3800, VIC, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, 3800, VIC, Australia.
| |
Collapse
|
67
|
Serum podocalyxin is significantly increased in early-onset preeclampsia and may represent a novel marker of maternal endothelial cell dysfunction. J Hypertens 2017; 35:2287-2294. [DOI: 10.1097/hjh.0000000000001461] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
68
|
Pan T, He G, Chen M, Bao C, Chen Y, Liu G, Zhou M, Li S, Xu W, Liu X. Abnormal CYP11A1 gene expression induces excessive autophagy, contributing to the pathogenesis of preeclampsia. Oncotarget 2017; 8:89824-89836. [PMID: 29163791 PMCID: PMC5685712 DOI: 10.18632/oncotarget.21158] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/08/2017] [Indexed: 12/20/2022] Open
Abstract
Objective In this study, we investigated the exact mechanism by which excessive CYP11A1 expression impairs the placentation process and whether this causes preeclampsia (PE) in an in vivo model. Setting and Design In order to study CYP11A1 overexpression, BeWo cells were transfected with CYP11A1. Pregnenolone, progesterone, and testosterone levels were measured by enzyme linked immunosorbent assays, and levels of autophagy markers were quantified by western blotting and immunofluorescence. Trophoblastic cell invasion was assessed using transwell assays; BeWo cells were treated with testosterone and an androgen receptor (AR) inhibitor (flutamide) to elucidate the invasion mechanism. An adenovirus overexpression rat model was established to investigate CYP11A1 overexpression in vivo and the phenotype was examined. Furthermore, human placenta samples (n = 24) were used to determine whether PE patient placentas showed altered CYP11A1 and autophagy marker expression. Results BeWo cells overexpressing CYP11A1 had significantly increased levels of pregnenolone, progesterone, and testosterone. Additionally, the expression levels of autophagy markers in CYP11A1-overexpressing BeWo cells were significantly increased. Trophoblast invasion was significantly reduced in CYP11A1-overexpressing cells as well as in cells treated with high testosterone. This reduction could be significantly rescued when cells were pretreated with flutamide. Overexpression of CYP11A1 in rat pregnancies led to PE-like symptoms and an over-activation of the AR-mediated pathway in the placenta. Elevated expression of CYP11A1 and autophagy markers could also be detected in PE placenta samples. Conclusions These results suggest that abnormally high expression of CYP11A1 induces trophoblast autophagy and inhibits trophoblastic invasion, which is associated with the etiology of PE.
Collapse
Affiliation(s)
- Tianying Pan
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Guolin He
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Meng Chen
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Chenyi Bao
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yan Chen
- Joint Laboratory of Reproductive Medicine, Sichuan University-The Chinese University of Hong Kong, Chengdu 610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Guangyu Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Mi Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Shuying Li
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Wenming Xu
- Joint Laboratory of Reproductive Medicine, Sichuan University-The Chinese University of Hong Kong, Chengdu 610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Xinghui Liu
- Joint Laboratory of Reproductive Medicine, Sichuan University-The Chinese University of Hong Kong, Chengdu 610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
69
|
Nourollahpour Shiadeh M, Riahi SM, Adam I, Saber V, Behboodi Moghadam Z, Armon B, Spotin A, Nazari Kangavari H, Rostami A. Helicobacter pylori infection and risk of preeclampsia: a systematic review and meta-analysis. J Matern Fetal Neonatal Med 2017; 32:324-331. [DOI: 10.1080/14767058.2017.1378331] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Malihe Nourollahpour Shiadeh
- Department of Midwifery and Reproductive Health, Nursing and Midwifery School, Tehran University of Medical Sciences, Tehran, Iran
- Department of Midwifery and Reproductive Health, Nursing and Midwifery School, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Mohammad Riahi
- Faculty of Health, Birjand University of Medical Sciences, Birjand, Iran
- Department of Epidemiology, School of Public Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ishag Adam
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Vafa Saber
- Department of Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Behboodi Moghadam
- Department of Midwifery and Reproductive Health, Nursing and Midwifery School, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahram Armon
- Psychosis Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Adel Spotin
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajar Nazari Kangavari
- Department of Epidemiology, School of Public Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Rostami
- Infectious Diseases and Tropical Medicine Research Center, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
70
|
Romero R, Erez O, Hüttemann M, Maymon E, Panaitescu B, Conde-Agudelo A, Pacora P, Yoon BH, Grossman LI. Metformin, the aspirin of the 21st century: its role in gestational diabetes mellitus, prevention of preeclampsia and cancer, and the promotion of longevity. Am J Obstet Gynecol 2017; 217:282-302. [PMID: 28619690 DOI: 10.1016/j.ajog.2017.06.003] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 05/30/2017] [Accepted: 06/05/2017] [Indexed: 12/16/2022]
Abstract
Metformin is everywhere. Originally introduced in clinical practice as an antidiabetic agent, its role as a therapeutic agent is expanding to include treatment of prediabetes mellitus, gestational diabetes mellitus, and polycystic ovarian disease; more recently, experimental studies and observations in randomized clinical trials suggest that metformin could have a place in the treatment or prevention of preeclampsia. This article provides a brief overview of the history of metformin in the treatment of diabetes mellitus and reviews the results of metaanalyses of metformin in gestational diabetes mellitus as well as the treatment of obese, non-diabetic, pregnant women to prevent macrosomia. We highlight the results of a randomized clinical trial in which metformin administration in early pregnancy did not reduce the frequency of large-for-gestational-age infants (the primary endpoint) but did decrease the frequency of preeclampsia (a secondary endpoint). The mechanisms by which metformin may prevent preeclampsia include a reduction in the production of antiangiogenic factors (soluble vascular endothelial growth factor receptor-1 and soluble endoglin) and the improvement of endothelial dysfunction, probably through an effect on the mitochondria. Another potential mechanism whereby metformin may play a role in the prevention of preeclampsia is its ability to modify cellular homeostasis and energy disposition, mediated by rapamycin, a mechanistic target. Metformin has a molecular weight of 129 Daltons and therefore readily crosses the placenta. There is considerable evidence to suggest that this agent is safe during pregnancy. New literature on the role of metformin as a chemotherapeutic adjuvant in the prevention of cancer and in prolonging life and protecting against aging is reviewed briefly. Herein, we discuss the mechanisms of action and potential benefits of metformin.
Collapse
|
71
|
Moore LG. Measuring high-altitude adaptation. J Appl Physiol (1985) 2017; 123:1371-1385. [PMID: 28860167 DOI: 10.1152/japplphysiol.00321.2017] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 08/15/2017] [Accepted: 08/15/2017] [Indexed: 12/12/2022] Open
Abstract
High altitudes (>8,000 ft or 2,500 m) provide an experiment of nature for measuring adaptation and the physiological processes involved. Studies conducted over the past ~25 years in Andeans, Tibetans, and, less often, Ethiopians show varied but distinct O2 transport traits from those of acclimatized newcomers, providing indirect evidence for genetic adaptation to high altitude. Short-term (acclimatization, developmental) and long-term (genetic) responses to high altitude exhibit a temporal gradient such that, although all influence O2 content, the latter also improve O2 delivery and metabolism. Much has been learned concerning the underlying physiological processes, but additional studies are needed on the regulation of blood flow and O2 utilization. Direct evidence of genetic adaptation comes from single-nucleotide polymorphism (SNP)-based genome scans and whole genome sequencing studies that have identified gene regions acted upon by natural selection. Efforts have begun to understand the connections between the two with Andean studies on the genetic factors raising uterine blood flow, fetal growth, and susceptibility to Chronic Mountain Sickness and Tibetan studies on genes serving to lower hemoglobin and pulmonary arterial pressure. Critical for future studies will be the selection of phenotypes with demonstrable effects on reproductive success, the calculation of actual fitness costs, and greater inclusion of women among the subjects being studied. The well-characterized nature of the O2 transport system, the presence of multiple long-resident populations, and relevance for understanding hypoxic disorders in all persons underscore the importance of understanding how evolutionary adaptation to high altitude has occurred.NEW & NOTEWORTHY Variation in O2 transport characteristics among Andean, Tibetan, and, when available, Ethiopian high-altitude residents supports the existence of genetic adaptations that improve the distribution of blood flow to vital organs and the efficiency of O2 utilization. Genome scans and whole genome sequencing studies implicate a broad range of gene regions. Future studies are needed using phenotypes of clear relevance for reproductive success for determining the mechanisms by which naturally selected genes are acting.
Collapse
Affiliation(s)
- Lorna G Moore
- Division of Reproductive Sciences, Department of Obstetrics & Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
72
|
Erez O, Romero R, Maymon E, Chaemsaithong P, Done B, Pacora P, Panaitescu B, Chaiworapongsa T, Hassan SS, Tarca AL. The prediction of late-onset preeclampsia: Results from a longitudinal proteomics study. PLoS One 2017; 12:e0181468. [PMID: 28738067 PMCID: PMC5524331 DOI: 10.1371/journal.pone.0181468] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 06/30/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Late-onset preeclampsia is the most prevalent phenotype of this syndrome; nevertheless, only a few biomarkers for its early diagnosis have been reported. We sought to correct this deficiency using a high through-put proteomic platform. METHODS A case-control longitudinal study was conducted, including 90 patients with normal pregnancies and 76 patients with late-onset preeclampsia (diagnosed at ≥34 weeks of gestation). Maternal plasma samples were collected throughout gestation (normal pregnancy: 2-6 samples per patient, median of 2; late-onset preeclampsia: 2-6, median of 5). The abundance of 1,125 proteins was measured using an aptamers-based proteomics technique. Protein abundance in normal pregnancies was modeled using linear mixed-effects models to estimate mean abundance as a function of gestational age. Data was then expressed as multiples of-the-mean (MoM) values in normal pregnancies. Multi-marker prediction models were built using data from one of five gestational age intervals (8-16, 16.1-22, 22.1-28, 28.1-32, 32.1-36 weeks of gestation). The predictive performance of the best combination of proteins was compared to placental growth factor (PIGF) using bootstrap. RESULTS 1) At 8-16 weeks of gestation, the best prediction model included only one protein, matrix metalloproteinase 7 (MMP-7), that had a sensitivity of 69% at a false positive rate (FPR) of 20% (AUC = 0.76); 2) at 16.1-22 weeks of gestation, MMP-7 was the single best predictor of late-onset preeclampsia with a sensitivity of 70% at a FPR of 20% (AUC = 0.82); 3) after 22 weeks of gestation, PlGF was the best predictor of late-onset preeclampsia, identifying 1/3 to 1/2 of the patients destined to develop this syndrome (FPR = 20%); 4) 36 proteins were associated with late-onset preeclampsia in at least one interval of gestation (after adjustment for covariates); 5) several biological processes, such as positive regulation of vascular endothelial growth factor receptor signaling pathway, were perturbed; and 6) from 22.1 weeks of gestation onward, the set of proteins most predictive of severe preeclampsia was different from the set most predictive of the mild form of this syndrome. CONCLUSIONS Elevated MMP-7 early in gestation (8-22 weeks) and low PlGF later in gestation (after 22 weeks) are the strongest predictors for the subsequent development of late-onset preeclampsia, suggesting that the optimal identification of patients at risk may involve a two-step diagnostic process.
Collapse
Affiliation(s)
- Offer Erez
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Maternity Department “D” and Obstetrical Day Care Center, Division of Obstetrics and Gynecology, Soroka University Medical Center, School of Medicine, Faculty of Heath Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, United States of America
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
- * E-mail: (RR); (ALT)
| | - Eli Maymon
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Piya Chaemsaithong
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Bogdan Done
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Percy Pacora
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Bogdan Panaitescu
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Sonia S. Hassan
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Adi L. Tarca
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, United States of America
- * E-mail: (RR); (ALT)
| |
Collapse
|
73
|
Romero R, Erez O, Maymon E, Chaemsaithong P, Xu Z, Pacora P, Chaiworapongsa T, Done B, Hassan SS, Tarca AL. The maternal plasma proteome changes as a function of gestational age in normal pregnancy: a longitudinal study. Am J Obstet Gynecol 2017; 217:67.e1-67.e21. [PMID: 28263753 PMCID: PMC5813489 DOI: 10.1016/j.ajog.2017.02.037] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/10/2017] [Accepted: 02/23/2017] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Pregnancy is accompanied by dramatic physiological changes in maternal plasma proteins. Characterization of the maternal plasma proteome in normal pregnancy is an essential step for understanding changes to predict pregnancy outcome. The objective of this study was to describe maternal plasma proteins that change in abundance with advancing gestational age and determine biological processes that are perturbed in normal pregnancy. STUDY DESIGN A longitudinal study included 43 normal pregnancies that had a term delivery of an infant who was appropriate for gestational age without maternal or neonatal complications. For each pregnancy, 3 to 6 maternal plasma samples (median, 5) were profiled to measure the abundance of 1125 proteins using multiplex assays. Linear mixed-effects models with polynomial splines were used to model protein abundance as a function of gestational age, and the significance of the association was inferred via likelihood ratio tests. Proteins considered to be significantly changed were defined as having the following: (1) >1.5-fold change between 8 and 40 weeks of gestation; and (2) a false discovery rate-adjusted value of P < .1. Gene ontology enrichment analysis was used to identify biological processes overrepresented among the proteins that changed with advancing gestation. RESULTS The following results were found: (1) Ten percent (112 of 1125) of the profiled proteins changed in abundance as a function of gestational age; (2) of the 1125 proteins analyzed, glypican-3, sialic acid-binding immunoglobulin-type lectin-6, placental growth factor, C-C motif-28, carbonic anhydrase 6, prolactin, interleukin-1 receptor 4, dual-specificity mitogen-activated protein kinase 4, and pregnancy-associated plasma protein-A had more than a 5-fold change in abundance across gestation (these 9 proteins are known to be involved in a wide range of both physiological and pathological processes, such as growth regulation, embryogenesis, angiogenesis immunoregulation, inflammation etc); and (3) biological processes associated with protein changes in normal pregnancy included defense response, defense response to bacteria, proteolysis, and leukocyte migration (false discovery rate, 10%). CONCLUSION The plasma proteome of normal pregnancy demonstrates dramatic changes in both the magnitude of changes and the fraction of the proteins involved. Such information is important to understand the physiology of pregnancy and the development of biomarkers to differentiate normal vs abnormal pregnancy and determine the response to interventions.
Collapse
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI.
| | - Offer Erez
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Eli Maymon
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Piya Chaemsaithong
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Zhonghui Xu
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI
| | - Percy Pacora
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Bogdan Done
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI
| | - Sonia S Hassan
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Adi L Tarca
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI.
| |
Collapse
|
74
|
Stoikou M, Grimolizzi F, Giaglis S, Schäfer G, van Breda SV, Hoesli IM, Lapaire O, Huhn EA, Hasler P, Rossi SW, Hahn S. Gestational Diabetes Mellitus Is Associated with Altered Neutrophil Activity. Front Immunol 2017; 8:702. [PMID: 28659928 PMCID: PMC5469883 DOI: 10.3389/fimmu.2017.00702] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 05/31/2017] [Indexed: 01/13/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is a unique form of glucose intolerance, in that it is transient and solely occurs in pregnancy. Pregnancies with GDM are at high risk of developing preeclampsia (PE), a leading cause of fetal and maternal morbidity or mortality. Since PE is associated with excessive activation of circulatory neutrophils and occurrence of neutrophil extracellular traps (NETs) in affected placentae, we examined these features in cases with GDM, as this could be a feature linking the two conditions. Our data indicate that neutrophil activity is indeed altered in GDM, exhibiting pronounced activation and spontaneous generation of NETs by isolated neutrophils in in vitro culture. In this manner, GDM may similarly affect neutrophil behavior and NET formation as witnessed in other forms of diabetes, with the addition of the physiological changes mediated by pregnancy. Since circulatory TNF-α levels are elevated in cases with GDM, a feature also observed in this study, we examined whether this pro-inflammatory cytokine contributed to neutrophil activation. By using infliximab, a clinically utilized TNF-α antagonist, we observed that the pro-NETotic effect of GDM sera was significantly reduced. We also detected pronounced neutrophil infiltrates in placentae from GDM cases. The occurrence of NETs in these tissues is suggested by the extracellular co-localization of citrullinated histones and myeloperoxidase. In addition, elevated neutrophil elastase (NE) mRNA and active enzymatic protein were also detected in such placentae. This latter finding could be important in the context of previous studies in cancer or diabetes model systems, which indicated that NE liberated from infiltrating neutrophils enters surrounding cells, altering cell signaling by the degradation of IRS1. These findings could potentiate the underlying inflammatory response process in GDM and possibly open an avenue for the therapeutic interventions in gestational hyperglycemia.
Collapse
Affiliation(s)
- Maria Stoikou
- Department of Biomedicine, University of Basel, University Hospital of Basel, Basel, Switzerland
| | - Franco Grimolizzi
- Department of Biomedicine, University of Basel, University Hospital of Basel, Basel, Switzerland.,Department Clinical Sciences, Polytechnic University Marche, Ancona, Italy
| | - Stavros Giaglis
- Department of Biomedicine, University of Basel, University Hospital of Basel, Basel, Switzerland.,Department of Rheumatology, Kantonsspital Aarau, Aarau, Switzerland
| | - Günther Schäfer
- Department of Biomedicine, University of Basel, University Hospital of Basel, Basel, Switzerland
| | - Shane Vontelin van Breda
- Department of Biomedicine, University of Basel, University Hospital of Basel, Basel, Switzerland
| | | | - Olav Lapaire
- University Women's Hospital, University Hospital of Basel, Basel, Switzerland
| | - Evelyn A Huhn
- University Women's Hospital, University Hospital of Basel, Basel, Switzerland
| | - Paul Hasler
- Department of Rheumatology, Kantonsspital Aarau, Aarau, Switzerland
| | - Simona W Rossi
- Department of Biomedicine, University of Basel, University Hospital of Basel, Basel, Switzerland
| | - Sinuhe Hahn
- Department of Biomedicine, University of Basel, University Hospital of Basel, Basel, Switzerland
| |
Collapse
|
75
|
Čonka J, Konečná B, Lauková L, Vlková B, Celec P. Fetal DNA does not induce preeclampsia-like symptoms when delivered in late pregnancy in the mouse. Placenta 2017; 52:100-105. [DOI: 10.1016/j.placenta.2017.02.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 02/06/2017] [Accepted: 02/08/2017] [Indexed: 10/20/2022]
|
76
|
Soh MC, Nelson-Piercy C. Biomarkers for Adverse Pregnancy Outcomes in Rheumatic Diseases. Rheum Dis Clin North Am 2017; 43:201-214. [PMID: 28390563 DOI: 10.1016/j.rdc.2016.12.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Pregnancy is a delicate balance of angiogenic factors. Adverse pregnancy outcomes in the form of placental insufficiency occur when antiangiogenic factors predominate, which manifests as maternal-placental syndrome (MPS). Women with rheumatic disease are at increased risk of MPS. Endothelial damage from circulating antiangiogenic factors and other inflammatory molecules in combination with preexisting maternal vascular risk factors is the likely underlying pathophysiological process for MPS. It is likely that these changes persist, and additional "insults" from ongoing inflammation, medications, and disease damage contribute to the development of accelerated cardiovascular disease seen in young women with rheumatic disease.
Collapse
Affiliation(s)
- May Ching Soh
- Silver Star High-Risk Pregnancy Unit, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Headley Way, Oxford OX3 9DU, UK; de Sweit Obstetric Medicine Department, Queen Charlotte's & Chelsea Hospital, Imperial College Healthcare NHS Trust, Du Cane Road, London W12 0HS, UK; Women's Health Academic Centre, King's College London, St Thomas' Hospital, 10th Floor, North Wing, Westminster Bridge Road, London SE1 7EH, UK.
| | - Catherine Nelson-Piercy
- de Sweit Obstetric Medicine Department, Queen Charlotte's & Chelsea Hospital, Imperial College Healthcare NHS Trust, Du Cane Road, London W12 0HS, UK; Women's Health Academic Centre, King's College London, St Thomas' Hospital, 10th Floor, North Wing, Westminster Bridge Road, London SE1 7EH, UK
| |
Collapse
|
77
|
Labarrere CA, DiCarlo HL, Bammerlin E, Hardin JW, Kim YM, Chaemsaithong P, Haas DM, Kassab GS, Romero R. Failure of physiologic transformation of spiral arteries, endothelial and trophoblast cell activation, and acute atherosis in the basal plate of the placenta. Am J Obstet Gynecol 2017; 216:287.e1-287.e16. [PMID: 28034657 DOI: 10.1016/j.ajog.2016.12.029] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 12/20/2016] [Indexed: 01/03/2023]
Abstract
BACKGROUND Failure of physiologic transformation of spiral arteries has been reported in preeclampsia, fetal growth restriction, fetal death, and spontaneous preterm labor with intact or ruptured membranes. Spiral arteries with failure of physiologic transformation are prone to develop atherosclerotic-like lesions of atherosis. There are striking parallels between preeclampsia and atherosclerotic disease, and between lesions of atherosis and atherosclerosis. Endothelial activation, identified by intercellular adhesion molecule-1 expression, is present in atherosclerotic-like lesions of heart transplantation, and is considered a manifestation of rejection. Similarly, endothelial activation/dysfunction has been implicated in the pathophysiology of atherosclerosis and preeclampsia. Intercellular adhesion molecule-1-overexpressing-activated endothelial cells are more resistant to trophoblast displacement than nonactivated endothelium, and may contribute to shallow spiral artery trophoblastic invasion in obstetrical syndromes having failure of physiologic transformation. OBJECTIVE We sought to determine whether failure of spiral artery physiologic transformation was associated with activation of interstitial extravillous trophoblasts and/or spiral artery endothelium and presence of acute atherosis in the placental basal plate. STUDY DESIGN A cross-sectional study of 123 placentas (19-42 weeks' gestation) obtained from normal pregnancies (n = 22), preterm prelabor rupture of membranes (n = 26), preterm labor (n = 23), preeclampsia (n = 27), intrauterine fetal death (n = 15), and small for gestational age (n = 10) was performed. Failure of spiral artery physiologic transformation and presence of cell activation was determined using immunohistochemistry of placental basal plates containing a median of 4 (minimum: 1; maximum: 9) vessels per placenta. Endothelial/trophoblast cell activation was defined by the expression of intercellular adhesion molecule-1. Investigators examining microscopic sections were blinded to clinical diagnosis. Pairwise comparisons among placenta groups were performed with Fisher exact test and Wilcoxon rank sum test using a Bonferroni-adjusted level of significance (.025). RESULTS We found that 87% (94/108) of placentas having spiral arteries with failure of physiologic transformation (actin-positive and cytokeratin-negative) in the basal plate, and 0% (0/15) of placentas having only spiral arteries with complete physiologic transformation (cytokeratin-positive and actin-negative), had arterial endothelial and/or interstitial extravillous trophoblasts reactive with the intercellular adhesion molecule-1 activation marker (P < .001). A significant correlation (R2 = 0.84) was found between expression of spiral artery endothelial and interstitial extravillous trophoblast intercellular adhesion molecule-1 (P < .001) in activated placentas. Lesions of atherosis were found in 31.9% (30/94) of placentas with complete and/or partial failure of physiologic transformation of spiral arteries that were intercellular adhesion molecule-1-positive, in none of the 14 placentas with failure of physiologic transformation that were intercellular adhesion molecule-1-negative, and in none of the 15 placentas with complete spiral artery physiologic transformation without failure (P = .001). All placentas (30/30, 100%) with atherosis were identified in placentas having concomitant spiral artery endothelial and interstitial extravillous trophoblast activation. CONCLUSION Failure of spiral artery physiologic transformation in the placental basal plate is associated with interstitial extravillous trophoblast and arterial endothelial activation along with increased frequency of spiral artery atherosis. These findings may be used to improve the characterization of different disorders of the placental bed such as in refining the existing tools for the early prediction of risk for preterm, preeclamptic, and other abnormal pregnancies.
Collapse
Affiliation(s)
- Carlos A Labarrere
- CBL Partners for Life, Indianapolis, IN; California Medical Innovations Institute, San Diego, CA.
| | | | - Elaine Bammerlin
- Indiana University Health Methodist Research Institute, Indianapolis, IN
| | - James W Hardin
- Department of Epidemiology and Biostatistics, University of South Carolina, Columbia, SC
| | - Yeon M Kim
- Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Piya Chaemsaithong
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | | | | | - Roberto Romero
- Center for Molecular Medicine and Genetics, Detroit, MI; Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI
| |
Collapse
|
78
|
Romero R, Chaemsaithong P, Tarca AL, Korzeniewski SJ, Maymon E, Pacora P, Panaitescu B, Chaiyasit N, Dong Z, Erez O, Hassan SS, Chaiworapongsa T. Maternal plasma-soluble ST2 concentrations are elevated prior to the development of early and late onset preeclampsia - a longitudinal study. J Matern Fetal Neonatal Med 2017; 31:418-432. [PMID: 28114842 DOI: 10.1080/14767058.2017.1286319] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The objectives of this study were to determine (1) the longitudinal profile of plasma soluble ST2 (sST2) concentrations in patients with preeclampsia and those with uncomplicated pregnancies; (2) whether the changes in sST2 occur prior to the diagnosis of preeclampsia; and (3) the longitudinal sST2 profile of women with early or late preeclampsia. MATERIALS AND METHODS This longitudinal nested case-control study included singleton pregnancies in the following groups: (1) uncomplicated pregnancies (n = 160); and (2) those complicated by early (<34 weeks, n = 9) and late (≥34 weeks, n = 31) preeclampsia. sST2 concentrations were determined by enzyme-linked immunosorbent assays. Mixed-effects models were used for the longitudinal analysis. RESULTS (1) Plasma sST2 concentration profiles across gestation differed significantly among cases and controls (p < 0.0001); (2) women with early preeclampsia had higher mean sST2 concentrations than controls at >22 weeks of gestation; cases with late preeclampsia had higher mean concentrations at >33 weeks of gestation (both p < 0.05); and (3) these changes started approximately 6 weeks prior to clinical diagnosis. CONCLUSIONS Maternal plasma sST2 concentrations are elevated 6 weeks prior to the clinical diagnosis of preeclampsia. An increase in the maternal plasma concentration of sST2 may contribute to an exaggerated intravascular inflammatory response and/or the Th1/Th2 imbalance in some cases.
Collapse
Affiliation(s)
- Roberto Romero
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,b Department of Obstetrics and Gynecology , University of Michigan , Ann Arbor , MI , USA.,c Department of Epidemiology and Biostatistics , Michigan State University , East Lansing , MI , USA.,d Center for Molecular Medicine and Genetics , Wayne State University , Detroit , MI , USA
| | - Piya Chaemsaithong
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Adi L Tarca
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Steven J Korzeniewski
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,c Department of Epidemiology and Biostatistics , Michigan State University , East Lansing , MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Eli Maymon
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Percy Pacora
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Bogdan Panaitescu
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Noppadol Chaiyasit
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Zhong Dong
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Offer Erez
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Sonia S Hassan
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Tinnakorn Chaiworapongsa
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| |
Collapse
|
79
|
Romero R, Erez O, Maymon E, Pacora P. Is an episode of suspected preterm labor that subsequently leads to a term delivery benign? Am J Obstet Gynecol 2017; 216:89-94. [PMID: 28148450 DOI: 10.1016/j.ajog.2016.12.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 12/19/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI.
| | - Offer Erez
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Eli Maymon
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Percy Pacora
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| |
Collapse
|
80
|
Suzuki S. Gestational weight gain in Japanese women with preeclampsia. HYPERTENSION RESEARCH IN PREGNANCY 2017. [DOI: 10.14390/jsshp.hrp2017-008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Shunji Suzuki
- Department of Obstetrics and Gynecology, Japanese Red Cross Katsushika Maternity Hospital
| |
Collapse
|
81
|
Abstract
Introduction: Hypertension in pregnancy is one of the three factors of maternal mortality. Etiology of the disease is unknown, but the many factors contributing to the identification and control of it can be taken a step to prevent and reduce the symptoms of the disease. The purpose of this study was to determine the prevalence of preeclampsia (high-blood pressure) in different seasons of the year. Methods: The present retrospective cross-sectional study was conducted on more than 8,000 pregnant women visiting Assali specialized hospital from 2011 to 2013. Required data was collected through questionnaire checklist. The Chi-square test with multiple comparisons was used to compare the frequencies of pregnancy-induced hypertension (PIH) according to the month of year, and adjustment of multiplicity was conducted using Bonferroni's method. Student's t-test was used to compare the means of PIH prevalence rates. In all analyses, P < 0.05 was taken to indicate statistical significance. Results: In these 8000 woman admitted to labor, overall prevalence of PIH was 3.8 ± 0.6%. The prevalence rate of PIH was highest in the summer (4.5%) and lowest frequent in the winter (2.7%), respectively. In July, the prevalence rate was significantly higher than those for any other month (4.7%), and in March, it was lower prevalence than for any month (2.2%), respectively. Using the Chi-square test, a significant difference between the incidence of disease was observed in summer and winter (P < 0.001). Conclusion: The prevalence rate of PIH was higher for delivery in summer and early spring and lowest for winter delivery among Khorramabad women based on these results; it seems that changes in temperature and humidity in different seasons can affect preeclampsia, and preeclampsia increases with increasing frequency temperature.
Collapse
Affiliation(s)
- Fatemeh Janani
- Social Determinants of Health Research Center, Faculty of Nursing and Midwifery, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Farahnaz Changaee
- Social Determinants of Health Research Center, Faculty of Nursing and Midwifery, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
82
|
Chaiworapongsa T, Romero R, Whitten AE, Korzeniewski SJ, Chaemsaithong P, Hernandez-Andrade E, Yeo L, Hassan SS. The use of angiogenic biomarkers in maternal blood to identify which SGA fetuses will require a preterm delivery and mothers who will develop pre-eclampsia. J Matern Fetal Neonatal Med 2016; 29:1214-28. [PMID: 26303962 DOI: 10.3109/14767058.2015.1048431] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To determine (1) whether maternal plasma concentrations of angiogenic and anti-angiogenic factors can predict which mothers diagnosed with "suspected small for gestational age fetuses (sSGA)" will develop pre-eclampsia (PE) or require an indicated early preterm delivery (≤ 34 weeks of gestation); and (2) whether risk assessment performance is improved using these proteins in addition to clinical factors and Doppler parameters. METHODS This prospective cohort study included women with singleton pregnancies diagnosed with sSGA (estimated fetal weight <10th percentile) between 24 and 34 weeks of gestation (n = 314). Plasma concentrations of soluble vascular endothelial growth factor receptor-1 (sVEGFR-1), soluble endoglin (sEng) and placental growth factor (PlGF) were determined in maternal blood obtained at the time of diagnosis. Doppler velocimetry of the umbilical (Umb) and uterine (UT) arteries was performed. The outcomes were (1) subsequent development of PE; and (2) indicated preterm delivery at ≤ 34 weeks of gestation (excluding deliveries as a result of spontaneous preterm labor, preterm pre-labor rupture of membranes or chorioamnionitis). RESULTS (1) The prevalence of PE and indicated preterm delivery was 9.2% (n = 29/314) and 7.3% (n = 23/314), respectively; (2) the area under the receiver operating characteristic curve (AUC) for the identification of patients who developed PE and/or required indicated preterm delivery was greater than 80% for the UT artery pulsatility index (PI) z-score and each biochemical marker (including their ratios) except sVEGFR-1 MoM; (3) using cutoffs at a false positive rate of 15%, women with abnormal plasma concentrations of angiogenic/anti-angiogenic factors were 7-13 times more likely to develop PE, and 12-22 times more likely to require preterm delivery than those with normal plasma MoM concentrations of these factors; (4) sEng, PlGF, PIGF/sEng and PIGF/sVEGFR-1 ratios MoM, each contributed significant information about the risk of PE beyond that provided by clinical factors and/or Doppler parameters: women who had low MoM values for these biomarkers were at 5-9 times greater risk of developing PE than women who had normal values, adjusting for clinical factors and Doppler parameters (adjusted odds ratio for PlGF: 9.1, PlGF/sEng: 5.6); (5) the concentrations of sVEGFR-1 and PlGF/sVEGFR-1 ratio MoM, each contributed significant information about the risk of indicated preterm delivery beyond that provided by clinical factors and/or Doppler parameters: women who had abnormal values were at 8-9 times greater risk for indicated preterm delivery, adjusting for clinical factors and Doppler parameters; and (6) for a two-stage risk assessment (Umb artery Doppler followed by Ut artery Doppler plus biochemical markers), among women who had normal Umb artery Doppler velocimetry (n = 279), 21 (7.5%) developed PE and 11 (52%) of these women were identified by an abnormal UT artery Doppler mean PI z-score (>2SD): a combination of PlGF/sEng ratio MoM concentration and abnormal UT artery Doppler velocimetry increased the sensitivity of abnormal UT artery Doppler velocimetry to 76% (16/21) at a fixed false-positive rate of 10% (p = 0.06). CONCLUSION Angiogenic and anti-angiogenic factors measured in maternal blood between 24 and 34 weeks of gestation can identify the majority of mothers diagnosed with "suspected SGA" who subsequently developed PE or those who later required preterm delivery ≤ 34 weeks of gestation. Moreover, incorporation of these biochemical markers significantly improves risk assessment performance for these outcomes beyond that of clinical factors and uterine and umbilical artery Doppler velocimetry.
Collapse
|
83
|
Systematic Review of Micro-RNA Expression in Pre-Eclampsia Identifies a Number of Common Pathways Associated with the Disease. PLoS One 2016; 11:e0160808. [PMID: 27529341 PMCID: PMC4986940 DOI: 10.1371/journal.pone.0160808] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 07/24/2016] [Indexed: 01/08/2023] Open
Abstract
Background Pre-eclampsia (PE) is a complex, multi-systemic condition of pregnancy which greatly impacts maternal and perinatal morbidity and mortality. MicroRNAs (miRs) are differentially expressed in PE and may be important in helping to understand the condition and its pathogenesis. Methods Case-control studies investigating expression of miRs in PE were collected through a systematic literature search. Data was extracted and compared from 58 studies to identify the most promising miRs associated with PE pathogenesis and identify areas of methodology which could account for often conflicting results. Results Some of the most frequently differentially expressed miRs in PE include miR-210, miR-223 and miR-126/126* which associate strongly with the etiological domains of hypoxia, immunology and angiogenesis. Members of the miR-515 family belonging to the imprinted chromosome 19 miR cluster with putative roles in trophoblast invasion were also found to be differentially expressed. Certain miRs appear to associate with more severe forms of PE such as miR-210 and the immune-related miR-181a and miR-15 families. Patterns of miR expression may help pinpoint key pathways (e.g. IL-6/miR-223/STAT3) and aid in untangling the heterogeneous nature of PE. The detectable presence of many PE-associated miRs in antenatal circulatory samples suggests their usefulness as predictive biomarkers. Further progress in ascertaining the clinical value of miRs and in understanding how they might contribute to pathogenesis is predicated upon resolving current methodological challenges in studies. These include differences in diagnostic criteria, cohort characteristics, sampling technique, RNA isolation and platform-dependent variation in miR profiling. Conclusion Reviewing studies of PE-associated miRs has revealed their potential as informants of underlying target genes and pathways relating to PE pathogenesis. However, the incongruity in results across current studies hampers their capacity to be useful biomarkers of the condition.
Collapse
|
84
|
Mastrolia SA, Novack L, Thachil J, Rabinovich A, Pikovsky O, Klaitman V, Loverro G, Erez O. LMWH in the prevention of preeclampsia and fetal growth restriction in women without thrombophilia. A systematic review and meta-analysis. Thromb Haemost 2016; 116:868-878. [PMID: 27440387 DOI: 10.1160/th16-02-0169] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/17/2016] [Indexed: 12/18/2022]
Abstract
Placental mediated pregnancy complications such as preeclampsia and fetal growth restriction (FGR) are common, serious, and associated with increased morbidity and mortality. We conducted a systematic review and meta-analysis to determine the effect of treatment with low-molecular-weight heparins (LMWHs) for secondary prevention of these complications in non thrombophilic women. We searched the electronic databases PubMed, Scopus, and Cochrane Library for randomised controlled trials addressing this question. Five studies including 403 patients met the inclusion criteria, 68 developed preeclampsia and 118 FGR. The studies were very heterogeneous in terms of inclusion criteria, LMWH preparation, and dosage. Meta-analyses were performed using random-effect models. The overall use of LMWHs was associated with a risk reduction for preeclampsia (Relative risk (RR) 0.366; 95 % confidence interval (CI), 0.219-0.614) and FGR (RR 0.409; 95 % CI, 0.195-0.932) vs. no treatment. From the data available for analysis it appears that the use of Dalteparin is associated with a risk reduction for preeclampsia (p=0.002) and FGR (p<0.001); while Enoxaparin is associated with risk reduction for preeclampsia (p=0.013) but not for FGR (p=0.3). In spite of the small number of studies addressing the research question, and the high variability among them, our meta-analysis found a modest beneficial effect of LMWH for secondary prevention of preeclampsia and FGR. Further studies are needed to address these questions before a definite conclusion can be reached.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Offer Erez
- Prof. Offer Erez, MD, Acting Director Maternity Department D and Obstetrical Day care Unit, Department of Obstetrics and Gynecology, Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University of the Negev, P. O.Box 151, 84101, Beer Sheva, Israel, Tel.: +972 8 6400061, E-mail
| |
Collapse
|
85
|
Klein E, Schlembach D, Ramoni A, Langer E, Bahlmann F, Grill S, Schaffenrath H, van der Does R, Messinger D, Verhagen-Kamerbeek WDJ, Reim M, Hund M, Stepan H. Influence of the sFlt-1/PlGF Ratio on Clinical Decision-Making in Women with Suspected Preeclampsia. PLoS One 2016; 11:e0156013. [PMID: 27243815 PMCID: PMC4887119 DOI: 10.1371/journal.pone.0156013] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 05/09/2016] [Indexed: 12/03/2022] Open
Abstract
Objective To evaluate the influence of the soluble fms-like tyrosine kinase 1/placental growth factor ratio in physicians’ decision making in pregnant women with signs and symptoms of preeclampsia in routine clinical practice. Methods A multicenter, prospective, open, non-interventional study enrolled pregnant women presenting with preeclampsia signs and symptoms in several European perinatal care centers. Before the soluble fms-like tyrosine kinase 1/placental growth factor ratio result was known, physicians documented intended clinical procedures using an iPad® application (data locked/time stamped). After the result was available, clinical decisions were confirmed or revised and documented. An independent adjudication committee evaluated the appropriateness of decisions based on maternal/fetal outcomes. Clinician decision making with regard to hospitalization was the primary outcome. Results In 16.9% of mothers (20/118) the hospitalization decision was changed after knowledge of the ratio. In 13 women (11.0%), the initial decision to hospitalize was changed to no hospitalization. In seven women (5.9%) the revised decision was hospitalization. All revised decisions were considered appropriate by the panel of adjudicators (McNemar test; p < 0.0001). Conclusions The use of the soluble fms-like tyrosine kinase 1/placental growth factor test influenced clinical decision making towards appropriate hospitalization in a considerable proportion of women with suspected preeclampsia. This is the first study to demonstrate the impact of angiogenic biomarkers on decision making in a routine clinical practice.
Collapse
Affiliation(s)
- Evelyn Klein
- Department of Gynecology and Obstetrics, Klinikum Rechts der Isar, TU München, Munich, Germany
| | | | - Angela Ramoni
- Department of Obstetrics and Gynecology, University Hospital of Innsbruck, Innsbruck, Austria
| | - Elena Langer
- Department of Obstetrics, Leipzig University, Leipzig, Germany
| | - Franz Bahlmann
- Department of Obstetrics, Bürgerhospital, Frankfurt, Germany
| | - Sabine Grill
- Department of Gynecology and Obstetrics, Klinikum Rechts der Isar, TU München, Munich, Germany
| | - Helene Schaffenrath
- Department of Obstetrics and Gynecology, University Hospital of Innsbruck, Innsbruck, Austria
| | | | | | | | - Manfred Reim
- Medical and Scientific Affairs, Roche Diagnostics GmbH, Penzberg, Germany
| | - Martin Hund
- Medical and Scientific Affairs, Roche Diagnostics International Ltd, Rotkreuz, Switzerland
| | - Holger Stepan
- Department of Obstetrics, Leipzig University, Leipzig, Germany
- * E-mail:
| |
Collapse
|
86
|
Brownfoot FC, Tong S, Hannan NJ, Hastie R, Cannon P, Kaitu'u-Lino TJ. Effects of simvastatin, rosuvastatin and pravastatin on soluble fms-like tyrosine kinase 1 (sFlt-1) and soluble endoglin (sENG) secretion from human umbilical vein endothelial cells, primary trophoblast cells and placenta. BMC Pregnancy Childbirth 2016; 16:117. [PMID: 27207105 PMCID: PMC4874016 DOI: 10.1186/s12884-016-0902-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/13/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Preeclampsia is associated with the placental release of soluble fms-like tyrosine kinase 1 (sFlt-1) and soluble endoglin (sENG). These anti-angiogenic factors cause hypertension and multi-organ injury. Pravastatin decreases placental secretion of sFlt-1 in vitro and is currently being examined in clinical trials as a potential treatment for preeclampsia. However, it is possible that different classes of statins may be more potent at decreasing sFlt-1 secretion. We compared the relative potency of three different generations of statins on sFlt-1 and sENG secretion from human endothelial cells, trophoblast cells, and placenta explants. METHODS We performed functional experiments using primary human umbilical vein endothelial cells, trophoblast cells and preterm preeclamptic placental explants to assess the affect of simvastatin, rosuvastatin and pravastatin on sFlt-1 and sENG secretion and compared the relative potency of each statin at reducing these factors (Inhibitory Concentration 50). Furthermore we assessed the effect of each statin on the antioxidant and cytoprotective enzyme, heme-oxygenase 1. RESULTS All statins reduced sFlt-1 secretion from endothelial cells, trophoblasts and preterm preeclamptic placental explants. Simvastatin was the most potent inhibitor of sFlt-1 secretion from endothelial cells (IC 50 3.2 μM), trophoblast cells (IC 50 61.4 μM) and placental explants. Simvastatin was 28 times and 3 times more potent at reducing sFlt-1 secretion from endothelial cells and 85 times and 33 times more potent at reducing sFlt-1 secretion from trophoblast cells than pravastatin or rosuvastatin respectively. All statins increased sENG secretion from endothelial cells however did not change secretion from placental explants. While all statins up-regulated heme-oxygenase 1 in endothelial cells, only simvastatin up-regulated its expression in placenta from patients with preterm preeclampsia. CONCLUSION Simvastatin may be a more potent inhibitor of sFlt-1 secretion from endothelial cells, trophoblast cells and placenta from women with preterm preeclampsia than either pravastatin or rosuvastatin.
Collapse
Affiliation(s)
- Fiona C Brownfoot
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, 163 Studley Road, Heidelberg, 3084, Victoria, Australia.
| | - Stephen Tong
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, 163 Studley Road, Heidelberg, 3084, Victoria, Australia
| | - Natalie J Hannan
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, 163 Studley Road, Heidelberg, 3084, Victoria, Australia
| | - Roxanne Hastie
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, 163 Studley Road, Heidelberg, 3084, Victoria, Australia
| | - Ping Cannon
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, 163 Studley Road, Heidelberg, 3084, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, 163 Studley Road, Heidelberg, 3084, Victoria, Australia
| |
Collapse
|
87
|
Kuo CY, Eranki A, Placone JK, Rhodes KR, Aranda-Espinoza H, Fernandes R, Fisher JP, Kim PCW. Development of a 3D Printed, Bioengineered Placenta Model to Evaluate the Role of Trophoblast Migration in Preeclampsia. ACS Biomater Sci Eng 2016; 2:1817-1826. [DOI: 10.1021/acsbiomaterials.6b00031] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Che-Ying Kuo
- Fischell
Department of Bioengineering, University of Maryland College Park, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
- Sheikh
Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, 111 Michigan Avenue NW, Washington, D.C. 20010, United States
| | - Avinash Eranki
- Sheikh
Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, 111 Michigan Avenue NW, Washington, D.C. 20010, United States
| | - Jesse K. Placone
- Fischell
Department of Bioengineering, University of Maryland College Park, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
| | - Kelly R. Rhodes
- Fischell
Department of Bioengineering, University of Maryland College Park, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
| | - Helim Aranda-Espinoza
- Fischell
Department of Bioengineering, University of Maryland College Park, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
| | - Rohan Fernandes
- Sheikh
Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, 111 Michigan Avenue NW, Washington, D.C. 20010, United States
- School
of Medicine and Health Sciences, The George Washington University, 2121 I Street, Washington, D.C. 20052, United States
| | - John P. Fisher
- Fischell
Department of Bioengineering, University of Maryland College Park, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
- Sheikh
Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, 111 Michigan Avenue NW, Washington, D.C. 20010, United States
| | - Peter C. W. Kim
- Sheikh
Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, 111 Michigan Avenue NW, Washington, D.C. 20010, United States
- School
of Medicine and Health Sciences, The George Washington University, 2121 I Street, Washington, D.C. 20052, United States
| |
Collapse
|
88
|
Korzeniewski SJ, Romero R, Chaiworapongsa T, Chaemsaithong P, Kim CJ, Kim YM, Kim JS, Yoon BH, Hassan SS, Yeo L. Maternal plasma angiogenic index-1 (placental growth factor/soluble vascular endothelial growth factor receptor-1) is a biomarker for the burden of placental lesions consistent with uteroplacental underperfusion: a longitudinal case-cohort study. Am J Obstet Gynecol 2016; 214:629.e1-629.e17. [PMID: 26688491 DOI: 10.1016/j.ajog.2015.11.015] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 09/21/2015] [Accepted: 11/18/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Placental lesions consistent with maternal vascular underperfusion (MVU) are thought to be pathogenically linked to preeclampsia, small-for-gestational-age newborns, fetal death, and spontaneous preterm labor and delivery; yet, these lesions cannot be diagnosed antenatally. We previously reported that patients with such conditions and lesions have an abnormal profile of the angiogenic placental growth factor (PlGF) and antiangiogenic factors (eg, soluble vascular endothelial growth factor receptor [sVEGFR]-1). OBJECTIVE The objectives of this study were to: (1) examine the relationship between the maternal plasma PlGF/sVEGFR-1 concentration ratio (referred to herein as angiogenic index-1) and the burden of histologic placental features consistent with MVU; and (2) test the hypothesis that angiogenic index-1 can identify patients in the midtrimester who are destined to deliver before 34 weeks of gestation with multiple (ie, ≥3) histologic placental features consistent with MVU. STUDY DESIGN A 2-stage case-cohort sampling strategy was used to select participants from among 4006 women with singleton gestations enrolled from 2006 through 2010 in a longitudinal study. Maternal plasma angiogenic index-1 ratios were determined using enzyme-linked immunosorbent assays. Placentas underwent histologic examination according to standardized protocols by experienced pediatric pathologists who were blinded to clinical diagnoses and pregnancy outcomes. The diagnosis of lesions consistent with MVU was made using criteria proposed by the Perinatal Section of the Society for Pediatric Pathology. Weighted analyses were performed to reflect the parent cohort; "n*" is used to reflect weighted frequencies. RESULTS (1) Angiogenic index-1 (PlGF/sVEGFR-1) concentration ratios were determined in 7560 plasma samples collected from 1499 study participants; (2) the prevalence of lesions consistent with MVU was 21% (n* = 833.9/3904) and 27% (n* = 11.4/42.7) of women with ≥3 MVU lesions delivered before 34 weeks of gestation; (3) a low angiogenic index-1 (<2.5th quantile for gestational age) in maternal plasma samples obtained within 48 hours of delivery had a sensitivity of 73% (n* = 8.3/11.4; 95% confidence interval [CI], 47-98%), a specificity of 94% (n* = 3130.9/3316.2; 95% CI, 94-95%), a positive likelihood ratio of 12.2, and a negative likelihood ratio of 0.29 in the identification of patients who delivered placentas with ≥3 MVU lesions at <34 weeks; (4) prospectively, at 20-23 weeks of gestation, a maternal plasma concentration of angiogenic index-1 <2.5th quantile identified 70% (n* = 7.2/10.3; 95% CI, 42-98%) of patients who delivered placentas with ≥3 MVU lesions before 34 weeks (specificity, 97% [n* = 2831.3/2918; 95% CI, 96-98%]; positive likelihood ratio, 23; negative likelihood ratio, 0.31); and (5) among women without obstetrical complications who delivered at term, angiogenic index-1 was lower in women with than without placental lesions consistent with MVU (P < .05). CONCLUSION Maternal plasma angiogenic index-1 (PlGF/sVEGFR-1) is the first biomarker for the burden of placental lesions consistent with MVU. We propose that an accumulation of these lesions in placentas delivered before 34 weeks is a histologic counterpart of an antiangiogenic profile.
Collapse
|
89
|
Triunfo S, Crovetto F, Crispi F, Rodriguez-Sureda V, Dominguez C, Nadal A, Peguero A, Gratacos E, Figueras F. Association of first-trimester angiogenic factors with placental histological findings in late-onset preeclampsia. Placenta 2016; 42:44-50. [PMID: 27238713 DOI: 10.1016/j.placenta.2016.04.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/04/2016] [Accepted: 04/05/2016] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To explore in women with late-onset preeclampsia (PE) the association between maternal levels of angiogenic/antiangiogenic factors in the first trimester of pregnancy and histological findings attributable to placental underperfusion (PUP). METHODS A nested case-control cohort study was conducted in 73 women with pregnancies complicated by late-onset PE (>34 weeks at delivery) matched with controls. First trimester uterine artery Doppler (UtA); maternal levels of placental growth factor (PlGF) and soluble fms-like tyrosine kinase-1 (sFlt-1) were retrieved. Placentas were histologically evaluated using a hierarchical and standardized classification system. One-way ANOVA with linear polynomial contrast or linear-by-linear association test was performed to test the hypothesis of a linear association across study groups (controls, PE without PUP and PE with PUP). RESULTS In 54 (74%) placentas, 89 placental histological findings qualifying for PUP were found. Across study groups, significant values were observed in maternal levels of decreased PlGF (MoM values: 1.53, 1.41 and 1.37; p < 0.001), increased sFlt-1 (MoM values: 3.11, 3.11 and 3.22; p = 0.002), increased sFlt-1/PlGF ratio (MoM values: 2.3, 2.3 and 2.44; p < 0.001), abnormal UtA Doppler (MoM values: 1, 1.26 and 1.32; p < 0.001), and worse perinatal outcomes in terms of gestational age at delivery, cesarean section for not reassuring fetal status, birth weight and neonatal acidosis. DISCUSSION In late-onset PE an imbalance of circulating angiogenic and anti-angiogenic factors already present at 8-10 weeks of pregnancy was associated with histological findings reflecting placental insufficiency. An early first trimester screening by angiogenic factors might help to identify patients with placental involvement among late-onset PE cases. CONCLUSION In late-onset preeclampsia, first-trimester uterine Doppler and circulating levels of angiogenic/antiangiogenic factors are associated with placental underperfusion.
Collapse
Affiliation(s)
- Stefania Triunfo
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain.
| | - Francesca Crovetto
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain; Ca' Granda, Ospedale Maggiore Policlinico, Dipartimento Ostetricia e Ginecologia, Università degli Studi di Milano, Milan, Italy
| | - Fatima Crispi
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Victor Rodriguez-Sureda
- Biochemistry and Molecular Biology Research Centre for Nanomedicine, Hospital Univeritari Vall d'Hebron, Centre for Biomedical Research on Rare Disease (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Carmen Dominguez
- Biochemistry and Molecular Biology Research Centre for Nanomedicine, Hospital Univeritari Vall d'Hebron, Centre for Biomedical Research on Rare Disease (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Alfons Nadal
- Department of Pathology, Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Anna Peguero
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Eduard Gratacos
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Francesc Figueras
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| |
Collapse
|
90
|
Finn-Sell SL, Renshall LJ, Cowley EJ, Dilworth MR, Wareing M, Greenwood SL, Sibley CP, Cottrell EC. The atrial natriuretic peptide (ANP) knockout mouse does not exhibit the phenotypic features of pre-eclampsia or demonstrate fetal growth restriction. Placenta 2016; 42:25-7. [PMID: 27238710 PMCID: PMC4898207 DOI: 10.1016/j.placenta.2016.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/15/2016] [Accepted: 04/04/2016] [Indexed: 12/11/2022]
Abstract
The ANP knockout mouse is reported to exhibit pregnancy-associated hypertension, proteinuria and impaired placental trophoblast invasion and spiral artery remodeling, key features of pre-eclampsia (PE). We hypothesized that these mice may provide a relevant model of human PE with associated fetal growth restriction (FGR). Here, we investigated pregnancies of ANP wild type (ANP+/+), heterozygous (ANP+/-) and knockout (ANP−/-) mice. Maternal blood pressure did not differ between genotypes (E12.5, E17.5), and fetal weight (E18.5) was unaffected. Placental weight was greater in ANP−/− versus ANP+/+ mice. Therefore, in our hands, the ANP model does not express phenotypic features of PE with FGR. Mouse models of pre-eclampsia and fetal growth restriction are needed to test potential therapies. ANP knockout mice have previously been identified as a potential model of pre-eclampsia. We find that these mice do not exhibit pregnancy-associated hypertension, or fetal growth restriction. ANP knockout mice do not provide a robust model of pre-eclampsia or fetal growth restriction.
Collapse
Affiliation(s)
- Sarah L Finn-Sell
- Maternal and Fetal Health Research Centre, Institute of Human Development, The University of Manchester, Manchester Academic Health Science Centre, St. Mary's Hospital, Manchester, United Kingdom.
| | - Lewis J Renshall
- Maternal and Fetal Health Research Centre, Institute of Human Development, The University of Manchester, Manchester Academic Health Science Centre, St. Mary's Hospital, Manchester, United Kingdom
| | - Elizabeth J Cowley
- Maternal and Fetal Health Research Centre, Institute of Human Development, The University of Manchester, Manchester Academic Health Science Centre, St. Mary's Hospital, Manchester, United Kingdom
| | - Mark R Dilworth
- Maternal and Fetal Health Research Centre, Institute of Human Development, The University of Manchester, Manchester Academic Health Science Centre, St. Mary's Hospital, Manchester, United Kingdom
| | - Mark Wareing
- Maternal and Fetal Health Research Centre, Institute of Human Development, The University of Manchester, Manchester Academic Health Science Centre, St. Mary's Hospital, Manchester, United Kingdom
| | - Susan L Greenwood
- Maternal and Fetal Health Research Centre, Institute of Human Development, The University of Manchester, Manchester Academic Health Science Centre, St. Mary's Hospital, Manchester, United Kingdom
| | - Colin P Sibley
- Maternal and Fetal Health Research Centre, Institute of Human Development, The University of Manchester, Manchester Academic Health Science Centre, St. Mary's Hospital, Manchester, United Kingdom
| | - Elizabeth C Cottrell
- Maternal and Fetal Health Research Centre, Institute of Human Development, The University of Manchester, Manchester Academic Health Science Centre, St. Mary's Hospital, Manchester, United Kingdom
| |
Collapse
|
91
|
Mooney SS, Lee RM, Tong S, Brownfoot FC. Expectant management of severe preterm preeclampsia: a comparison of maternal and fetal indications for delivery. J Matern Fetal Neonatal Med 2016; 29:3821-6. [DOI: 10.3109/14767058.2016.1147555] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Samantha S. Mooney
- Department of Obstetrics and Gynaecology, Mercy Hospital for Women, Heidelberg, Victoria, Australia and
| | - Rilka M. Lee
- Department of Obstetrics and Gynaecology, Mercy Hospital for Women, Heidelberg, Victoria, Australia and
| | - Stephen Tong
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Fiona C. Brownfoot
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| |
Collapse
|
92
|
Metformin as a prevention and treatment for preeclampsia: effects on soluble fms-like tyrosine kinase 1 and soluble endoglin secretion and endothelial dysfunction. Am J Obstet Gynecol 2016; 214:356.e1-356.e15. [PMID: 26721779 DOI: 10.1016/j.ajog.2015.12.019] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 12/15/2015] [Accepted: 12/15/2015] [Indexed: 01/28/2023]
Abstract
BACKGROUND Preeclampsia is associated with placental ischemia/hypoxia and secretion of soluble fms-like tyrosine kinase 1 and soluble endoglin into the maternal circulation. This causes widespread endothelial dysfunction that manifests clinically as hypertension and multisystem organ injury. Recently, small molecule inhibitors of hypoxic inducible factor 1α have been found to reduce soluble fms-like tyrosine kinase 1 and soluble endoglin secretion. However, their safety profile in pregnancy is unknown. Metformin is safe in pregnancy and is also reported to inhibit hypoxic inducible factor 1α by reducing mitochondrial electron transport chain activity. OBJECTIVE The purposes of this study were to determine (1) the effects of metformin on placental soluble fms-like tyrosine kinase 1 and soluble endoglin secretion, (2) to investigate whether the effects of metformin on soluble fms-like tyrosine kinase 1 and soluble endoglin secretion are regulated through the mitochondrial electron transport chain, and (3) to examine its effects on endothelial dysfunction, maternal blood vessel vasodilation, and angiogenesis. STUDY DESIGN We performed functional (in vitro and ex vivo) experiments using primary human tissues to examine the effects of metformin on soluble fms-like tyrosine kinase 1 and soluble endoglin secretion from placenta, endothelial cells, and placental villous explants. We used succinate, mitochondrial complex II substrate, to examine whether the effects of metformin on soluble fms-like tyrosine kinase 1 and soluble endoglin secretion were mediated through the mitochondria. We also isolated mitochondria from preterm preeclamptic placentas and gestationally matched control subjects and measured mitochondrial electron transport chain activity using kinetic spectrophotometric assays. Endothelial cells or whole maternal vessels were incubated with metformin to determine whether it rescued endothelial dysfunction induced by either tumor necrosis factor-α (to endothelial cells) or placenta villous explant-conditioned media (to whole vessels). Finally, we examined the effects of metformin on angiogenesis on maternal omental vessel explants. RESULTS Metformin reduced soluble fms-like tyrosine kinase 1 and soluble endoglin secretion from primary endothelial cells, villous cytotrophoblast cells, and preterm preeclamptic placental villous explants. The reduction in soluble fms-like tyrosine kinase 1 and soluble endoglin secretion was rescued by coadministration of succinate, which suggests that the effects of metformin on soluble fms-like tyrosine kinase 1 and soluble endoglin were likely to be regulated at the level of the mitochondria. In addition, the mitochondrial electron transport chain inhibitors rotenone and antimycin reduced soluble fms-like tyrosine kinase 1 secretion, which further suggests that soluble fms-like tyrosine kinase 1 secretion is regulated through the mitochondria. Mitochondrial electron transport chain activity in preterm preeclamptic placentas was increased compared with gestation-matched control subjects. Metformin improved features of endothelial dysfunction relevant to preeclampsia. It reduced endothelial cell messenger RNA expression of vascular cell adhesion molecule 1 that was induced by tumor necrosis factor-α (vascular cell adhesion molecule 1 is an inflammatory adhesion molecule up-regulated with endothelial dysfunction and is increased in preeclampsia). Placental conditioned media impaired bradykinin-induced vasodilation; this effect was reversed by metformin. Metformin also improved whole blood vessel angiogenesis impaired by fms-like tyrosine kinase 1. CONCLUSION Metformin reduced soluble fms-like tyrosine kinase 1 and soluble endoglin secretion from primary human tissues, possibly by inhibiting the mitochondrial electron transport chain. The activity of the mitochondrial electron transport chain was increased in preterm preeclamptic placenta. Metformin reduced endothelial dysfunction, enhanced vasodilation in omental arteries, and induced angiogenesis. Metformin has potential to prevent or treat preeclampsia.
Collapse
|
93
|
Abstract
PURPOSE OF REVIEW Hypertension in pregnancy contributes substantially to perinatal mortality and morbidity of both the mother and her child. High blood pressure is mainly responsible for this adverse outcome, in particular when associated with preeclampsia. Although preeclampsia is nowadays a well-known clinical-obstetrical entity, and screening for this complication has been part of routine care during pregnancy for nearly 100 years, its cause is still enigmatic. RECENT FINDINGS Profound changes of the demographic development of our society, the worldwide rising prevalence of obesity and metabolic disorders, and progress in reproductive medicine will inevitably modify the prevalence of many medical problems in pregnancy. Complications such as gestational diabetes mellitus, chronic hypertension, and preeclampsia will rise and an interdisciplinary approach is necessary to handle these women during pregnancy and also after delivery. Indeed, it is now well established that these women and their offspring born large or small-for-gestational age are at increased risk for severe cardiovascular and metabolic complications later in life. SUMMARY Knowledge of the pregnancy course is not only important for an obstetrician but also increasingly inevitable for the general practitioner. Recognition, classification, and adequate management of hypertensive pregnancy disorders and associated complications may considerably reduce perinatal death and morbidity.
Collapse
|
94
|
Kirbas A, Ersoy AO, Daglar K, Dikici T, Biberoglu EH, Kirbas O, Danisman N. Prediction of Preeclampsia by First Trimester Combined Test and Simple Complete Blood Count Parameters. J Clin Diagn Res 2015; 9:QC20-3. [PMID: 26674673 DOI: 10.7860/jcdr/2015/15397.6833] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 09/10/2015] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Preeclampsia is a serious disease which may result in maternal and neonatal mortality and morbidity. Improving the outcome for preeclampsia necessitates early prediction of the disease to identify women at high risk. Measuring blood cell subtype ratios, such as the neutrophil to lymphocyte (NLR) and platelet to lymphocyte (PLR) ratios, might provide prognostic and diagnostic clues to diseases. AIM To investigate hematological changes in early pregnancy, using simple complete blood count (CBC) and blood concentrations of beta-human chorionic gonadotropin (β-hCG) and pregnancy-associated plasma protein-A (PAPP-A) to determine whether these measures are of any value in the prediction and early diagnosis of preeclampsia. MATERIALS AND METHODS Six hundred fourteen consecutive pregnant women with preeclampsia (288 with mild disease and 326 with severe disease) and 320 uncomplicated pregnant women were included in the study. Blood samples for routine CBC and first trimester screen, which combines PAPP-A and free β-hCG blood concentrations, were analyzed. RESULTS The NLR values were significantly higher in the severe preeclampsia group compared with the control group (p<0.001). We also confirmed that levels of PAPP-A were lower in patients who developed preeclampsia. CONCLUSION Because measuring CBC parameters, particularly NLR, is fast and easily applicable, they may be used to predict preeclampsia.
Collapse
Affiliation(s)
- Ayse Kirbas
- Faculty, Department of Perinatology, Zekai Tahir Burak Women's Health Education and Research Hospital , Ankara, Turkey
| | - Ali Ozgur Ersoy
- Faculty, Department of Perinatology, Zekai Tahir Burak Women's Health Education and Research Hospital , Ankara, Turkey
| | - Korkut Daglar
- Faculty, Department of Perinatology, Zekai Tahir Burak Women's Health Education and Research Hospital , Ankara, Turkey
| | - Turkan Dikici
- Faculty, Deparment of Obstetric and Gynecology, Hilal Hospital , Kirikkale, Turkey
| | - Ebru Hacer Biberoglu
- Faculty, Department of Perinatology, Zekai Tahir Burak Women's Health Education and Research Hospital , Ankara, Turkey
| | - Ozgur Kirbas
- Faculty, Department of Cardiology, Yuksek Ihtisas Education and Research Hospital , Ankara, Turkey
| | - Nuri Danisman
- Assistant Professor, Department of Perinatology, Zekai Tahir Burak Women's Health Education and Research Hospital , Ankara, Turkey
| |
Collapse
|
95
|
Martin E, Ray PD, Smeester L, Grace MR, Boggess K, Fry RC. Epigenetics and Preeclampsia: Defining Functional Epimutations in the Preeclamptic Placenta Related to the TGF-β Pathway. PLoS One 2015; 10:e0141294. [PMID: 26510177 PMCID: PMC4624949 DOI: 10.1371/journal.pone.0141294] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/07/2015] [Indexed: 12/19/2022] Open
Abstract
Preeclampsia is a potentially fatal pregnancy disorder affecting millions of women around the globe. Dysregulation in gene and protein expression within key biological pathways controlling angiogenesis has been implicated in the development of preeclampsia. Altered CpG methylation, a type of epimutation, may underlie this pathway dysregulation. In the present study, placental tissue from preeclamptic cases and normotensive controls was analyzed for genome-wide differential CpG methylation and concomitant changes in gene expression. A set of 123 genes, representing 19.9% of all genes with altered CpG methylation, was associated with functional changes in transcript levels. Underscoring the complex relationships between CpG methylation and gene expression, here hypermethylation was never associated with gene silencing, nor was hypomethylation always associated with gene activation. Moreover, the genomic region of the CpG mark was important in predicting the relationship between CpG methylation and gene expression. The 123 genes were enriched for their involvement in the transforming growth factor beta (TGF-β) signaling pathway, a known regulator of placental trophoblast invasion and migration. This is the first study to identify CpG hypomethylation as an activator of TGF-β-associated gene expression in the preeclamptic placenta. The results suggest functional epimutations are associated with preeclampsia disease status and the identified genes may represent novel biomarkers of disease.
Collapse
Affiliation(s)
- Elizabeth Martin
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, 135 Dauer Drive, CB 7431, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Paul D. Ray
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, 135 Dauer Drive, CB 7431, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Curriculum in Toxicology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Lisa Smeester
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, 135 Dauer Drive, CB 7431, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Matthew R. Grace
- Department of Obstetrics & Gynecology, University of North Carolina School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Kim Boggess
- Department of Obstetrics & Gynecology, University of North Carolina School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Rebecca C. Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, 135 Dauer Drive, CB 7431, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Curriculum in Toxicology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
96
|
Brownfoot FC, Tong S, Hannan NJ, Hastie R, Cannon P, Tuohey L, Kaitu'u-Lino TJ. YC-1 reduces placental sFlt-1 and soluble endoglin production and decreases endothelial dysfunction: A possible therapeutic for preeclampsia. Mol Cell Endocrinol 2015; 413:202-8. [PMID: 26159901 DOI: 10.1016/j.mce.2015.06.033] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 06/05/2015] [Accepted: 06/28/2015] [Indexed: 01/12/2023]
Abstract
Preeclampsia is a serious complication of pregnancy with no medical treatment. It is caused by intermittent placental hypoxia and release of sFlt-1 and soluble endoglin, leading to wide spread maternal endothelial dysfunction and multisystem organ injury. YC-1 is a guanylyl cyclase activator and HIF1α inhibitor developed for use in hypertension and atherosclerosis. We examined whether YC-1 reduces sFlt-1 and sENG secretion and reverses endothelial dysfunction in primary human tissues. YC-1 significantly reduced sFlt-1 and sENG secretion from human umbilical vein endothelial cells, purified primary trophoblast cells and placental explants taken from patients with preterm preeclampsia. This was concordant with reduced HIF1α expression. YC-1 also reversed TNFα induced endothelial dysfunction, including reduced vascular cell adhesion molecule 1 expression and monocyte adhesion to primary endothelial cells. We conclude YC-1 decreases placental production of sFlt-1 and sENG and decreases endothelial dysfunction. It is a novel therapeutic candidate for preeclampsia.
Collapse
Affiliation(s)
- Fiona C Brownfoot
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Rd, Heidelberg 3084, Victoria, Australia.
| | - Stephen Tong
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Rd, Heidelberg 3084, Victoria, Australia
| | - Natalie J Hannan
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Rd, Heidelberg 3084, Victoria, Australia
| | - Roxanne Hastie
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Rd, Heidelberg 3084, Victoria, Australia
| | - Ping Cannon
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Rd, Heidelberg 3084, Victoria, Australia
| | - Laura Tuohey
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Rd, Heidelberg 3084, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Rd, Heidelberg 3084, Victoria, Australia
| |
Collapse
|
97
|
Ferraro ZM, Contador F, Tawfiq A, Adamo KB, Gaudet L. Gestational weight gain and medical outcomes of pregnancy. Obstet Med 2015; 8:133-7. [PMID: 27512468 DOI: 10.1177/1753495x15591320] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
This narrative review discusses gestational weight gain (GWG) and medical outcomes of pregnancy, including metabolic, cardiovascular, respiratory, musculoskeletal and psychiatric systems. Taken as a whole, the available evidence shows that excessive GWG increases the risk of all medical complications of pregnancy, and negatively impacts the long-term health and weight of both mothers and their offspring. Briefly, interventions to encourage appropriate GWG are discussed and readers are directed to resources to facilitate discussion of pregnancy weight.
Collapse
Affiliation(s)
- Zachary M Ferraro
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Newborn Care, The Ottawa Hospital, Ottawa, Canada; Healthy Active Living and Obesity (HALO) Research Group, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada; Ottawa Hospital Research Institute, Ottawa, Canada
| | - Fernanda Contador
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Newborn Care, The Ottawa Hospital, Ottawa, Canada
| | - Afaf Tawfiq
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Newborn Care, The Ottawa Hospital, Ottawa, Canada
| | - Kristi B Adamo
- Healthy Active Living and Obesity (HALO) Research Group, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Laura Gaudet
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Newborn Care, The Ottawa Hospital, Ottawa, Canada; Ottawa Hospital Research Institute, Ottawa, Canada; Faculty of Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
98
|
Hahn S, Lapaire O, Than NG. Biomarker development for presymptomatic molecular diagnosis of preeclampsia: feasible, useful or even unnecessary? Expert Rev Mol Diagn 2015; 15:617-29. [PMID: 25774007 PMCID: PMC4673513 DOI: 10.1586/14737159.2015.1025757] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The past decade saw the advent of a number of promising biomarkers to detect pregnancies at risk for preeclampsia (PE), the foremost being those associated with an imbalance of angiogenic factors. In late pregnancy, these are useful for the detection of imminent cases of PE, while earlier they were more predictive for early- than late-onset PE. This suggests that there may be fundamental differences between the underlying pathology of these two PE forms. Therefore, it is possible that such a biological premise may limit the development of biomarkers that will permit the efficacious detection of both early- and late-onset PE via an analysis of first-trimester maternal blood samples. Consequently, a significant increase in our understanding of the underlying pathology of PE, using a variety of approaches ranging from systems biology to animal models, will be necessary in order to overcome this obstacle.
Collapse
Affiliation(s)
- Sinuhe Hahn
- Laboratory for Prenatal Medicine, Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, CH 4031 Basel, Switzerland
| | | | | |
Collapse
|
99
|
MiR-519d-3p suppresses invasion and migration of trophoblast cells via targeting MMP-2. PLoS One 2015; 10:e0120321. [PMID: 25803859 PMCID: PMC4372600 DOI: 10.1371/journal.pone.0120321] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 02/08/2015] [Indexed: 11/19/2022] Open
Abstract
Our study was approved by the Medical Ethics Committee of Tang Du Hospital, Fourth Military Medical University and complied strictly with national ethical guidelines. Preeclampsia (PE) is a specific clinical disorder characterized by gestational hypertension and proteinuria and is a leading cause of maternal and perinatal mortality worldwide. The miR-519d-3p is upregulated in the maternal plasma of patients with PE which indicates a possible association between this microRNA and the pathogenesis of PE. No studies to date have addressed the effect of miR-519d-3p on the invasion and migration of trophoblast cells. In our study, we found that miR-519d-3p expression was elevated in placental samples from patients with PE. In vitro, overexpression of miR-519d-3p significantly inhibited trophoblast cell migration and invasion, whereas transfection of a miR-519d-3p inhibitor enhanced trophoblast cell migration and invasion. Luciferase assays confirmed that matrix metalloproteinase-2 (MMP-2) is a direct target of miR-519d-3p. Quantitative real-time PCR and western blot assays showed that overexpression of miR-519d-3p downregulated MMP-2 mRNA and protein expression. Knockdown of MMP-2 using a siRNA attenuated the increased trophoblast migration and invasion promoted by the miR-519d-3p inhibitor. In placentas from patients with PE or normal pregnancies, a negative correlation between the expression of MMP-2 and miR-519d-3p was observed using the Pearson correlation and linear regression analysis. Our present findings suggest that upregulation of miR-519d-3p may contribute to the development of PE by inhibiting trophoblast cell migration and invasion via targeting MMP-2; miR-519d-3p may represent a potential predictive and therapeutic target for PE.
Collapse
|
100
|
Zhu XL, Wang J, Jiang RZ, Teng YC. Pulsatility index in combination with biomarkers or mean arterial pressure for the prediction of pre-eclampsia: Systematic literature review and meta-analysis. Ann Med 2015; 47:414-22. [PMID: 26153822 DOI: 10.3109/07853890.2015.1059483] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Our objective was to perform a meta-analysis examining the sensitivity of pulsatility index (PI) and various biomarkers and PI and mean arterial pressure (MAP) for the prediction of pre-eclampsia. MATERIAL AND METHODS PubMed, CENTRAL, and Embase databases were searched from inception until 8 May 2014 using combinations of the search terms: pre-eclampsia, ultrasonography, pregnancy, biomarker, mean arterial pressure, placental protein 13, pregnancy-associated plasma protein-A, placental growth factor, activin A, inhibin A, pulsatility index. The pooled sensitivity of PI + biomarkers and PI + MAP were calculated, and reported with corresponding 95% confidence intervals (CIs). RESULTS Fifteen studies were included in the meta-analysis. The pooled sensitivity of all biomarkers for the prediction of pre-eclampsia was 0.669 (95% CI 0.610-0.723), for the prediction of early-onset pre-eclampsia was 0.830 (95% CI 0.794-0.861), and for the prediction of late-onset pre-eclampsia was 0.564 (95% CI 0.499-0.627). Similarly, the predictive ability of PI + MAP for early-onset pre-eclampsia was good (sensitivity 0.894), while that for late-onset was poor (sensitivity 0.570). CONCLUSION The combination of PI and different biomarkers or MAP exhibits a good predictive ability for early-onset pre-eclampsia, and poor predictive ability for late-onset pre-eclampsia.
Collapse
Affiliation(s)
- Xiao-Lu Zhu
- a Department of Obstetrics and Gynecology , Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai 200233 , P.R. China
| | | | | | | |
Collapse
|