951
|
Navaratnam K, Abreu P, Clarke H, Jorgensen A, Alfirevic A, Alfirevic Z. Evaluation of agreement of placental growth factor (PlGF) tests and the soluble FMS-like tyrosine kinase 1 (sFlt-1)/PlGF ratio, comparison of predictive accuracy for pre-eclampsia, and relation to uterine artery Doppler and response to aspirin. J Matern Fetal Neonatal Med 2017; 32:179-187. [PMID: 28851242 DOI: 10.1080/14767058.2017.1373760] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVES The objective of this study is to evaluate agreement between PlGF and sFlt-1/PlGF ratio tests and compare their predictive accuracy for pre-eclampsia in high-risk women. Also, to examine for associations of abnormal PlGF or sFlt-1/PlGF ratio with abnormal uterine artery Doppler and platelet response to aspirin. METHODS Prospective cohort study, 150 pregnant women at high risk of pre-eclampsia prescribed 75 mg aspirin daily. Uterine artery Dopplers were assessed at 20+0-23+6 weeks. At 33+0-35+6 weeks platelet function aspirin metabolites, PlGF and the sFlt-1/PlGF ratio were measured. OUTCOME Measures were all pre-eclampsia and pre-eclampsia requiring delivery prior to 37 weeks. RESULTS Overall percent agreement was 89.3% for PlGF tests but 74.7-78% for PlGF tests and the sFlt-1/PlGF ratio. AUCs were 0.70-0.75 for prediction of any pre-eclampsia and 0.92-0.99 for preterm pre-eclampsia. We found a significant association between abnormal PlGF or sFlt-1/PlGF ratio and abnormal uterine artery Doppler (χ2 5.47, p = .019), but no association with platelet response to aspirin (χ2 0.12, p = .913). There were no associations between suboptimal aspirin adherence and either abnormal angiogenic markers or uterine artery Dopplers (χ2 0.144, 0.038, p = .704, .846, respectively). CONCLUSIONS There was good agreement between PlGF tests and limited agreement between PlGF tests and the sFlt-1/PlGF ratio. All tests have heightened predictive accuracy for preterm pre-eclampsia. Abnormal PlGF or sFlt-1/PlGF ratio relates to abnormal uterine artery Doppler but not platelet response to aspirin.
Collapse
Affiliation(s)
- Kate Navaratnam
- a Centre for Women's Health Research, Institute of Translational Medicine , University of Liverpool , Liverpool , UK.,b Liverpool Women's Hospital , Liverpool , UK
| | | | | | - Andrea Jorgensen
- c Department of Biostatistics, Institute of Translational Medicine , University of Liverpool , Liverpool , UK
| | - Ana Alfirevic
- d The Wolfson Centre for Personalised Medicine, Institute of Translational Medicine , University of Liverpool , Liverpool , UK
| | - Zarko Alfirevic
- a Centre for Women's Health Research, Institute of Translational Medicine , University of Liverpool , Liverpool , UK.,b Liverpool Women's Hospital , Liverpool , UK
| |
Collapse
|
952
|
Grand'Maison S, Pilote L, Schlosser K, Stewart DJ, Okano M, Dayan N. Clinical Features and Outcomes of Acute Coronary Syndrome in Women With Previous Pregnancy Complications. Can J Cardiol 2017; 33:1683-1692. [PMID: 29173607 DOI: 10.1016/j.cjca.2017.08.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/18/2017] [Accepted: 08/18/2017] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Women with previous cardiometabolic complications of pregnancy experience double the risk of cardiovascular disease. However, few data exist on the clinical effect of these complications at the time of an acute coronary syndrome (ACS). The objective of this work was to compare risk factors, clinical features, and outcomes among women with premature ACS with or without previous pregnancy complications (gestational diabetes and/or hypertensive disorders of pregnancy). METHODS Data were obtained from a multicentre cohort of individuals hospitalized with premature ACS. A total of 251 parous women were included and provided obstetric history and blood samples. They were followed for the development of major adverse cardiac events at 12 months. RESULTS At presentation with ACS, women with a previous pregnancy complication (38%) were slightly younger than were women without such complications (47.4 ± 6.2 vs 49.1 ± 5.6 years; P = 0.002). They also had more traditional atherosclerotic risk factors. Specifically, women with previous preeclampsia were more likely to have chronic hypertension and an elevated ratio of soluble fms-like tyrosine kinase:placental growth factor. There was no between-group difference in Global Registry of Acute Coronary Events (GRACE) score or troponin tertile but there was a trend toward higher risk of ST-elevation myocardial infarction in women who had a previous pregnancy complication (odds ratio, 1.80; 95% confidence interval, 1.00-3.23; P = 0.05). There was also an increased risk of recurrent ACS at 12 months in women with previous preeclampsia (hazard ratio, 6.79; 95% confidence interval, 1.37-33.63; P = 0.02). CONCLUSIONS Among a cohort of women with ACS, previous pregnancy complications were associated with more severe disease and poorer outcome.
Collapse
Affiliation(s)
- Sophie Grand'Maison
- Division of General Internal Medicine, Centre hospitalier de l'Université de Montréal, Montréal, Québec, Canada; Centre de Recherche du Centre hospitalier de l'Université de Montréal, Montreal, Québec, Canada
| | - Louise Pilote
- Research Institute, McGill University Health Centre, Montreal, Quebec, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada; Division of General Internal Medicine, McGill University Health Centre, Montreal, Quebec, Canada.
| | - Kenny Schlosser
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Duncan J Stewart
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Marisa Okano
- Research Institute, McGill University Health Centre, Montreal, Quebec, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - Natalie Dayan
- Research Institute, McGill University Health Centre, Montreal, Quebec, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada; Division of General Internal Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
953
|
First-trimester placental thickness and the risk of preeclampsia or SGA. Placenta 2017; 57:123-128. [DOI: 10.1016/j.placenta.2017.06.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 06/13/2017] [Accepted: 06/17/2017] [Indexed: 01/02/2023]
|
954
|
Autophagy regulation in preeclampsia: Pros and cons. J Reprod Immunol 2017; 123:17-23. [DOI: 10.1016/j.jri.2017.08.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 08/16/2017] [Indexed: 12/11/2022]
|
955
|
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: 161] [Impact Index Per Article: 20.1] [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
|
956
|
Perales A, Delgado JL, de la Calle M, García‐Hernández JA, Escudero AI, Campillos JM, Sarabia MD, Laíz B, Duque M, Navarro M, Calmarza P, Hund M, Álvarez FV. sFlt-1/PlGF for prediction of early-onset pre-eclampsia: STEPS (Study of Early Pre-eclampsia in Spain). ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2017; 50:373-382. [PMID: 27883242 PMCID: PMC5836987 DOI: 10.1002/uog.17373] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 11/08/2016] [Accepted: 11/11/2016] [Indexed: 05/31/2023]
Abstract
OBJECTIVE A high ratio of soluble fms-like tyrosine kinase-1 (sFlt-1) to placental growth factor (PlGF) has been linked to pre-eclampsia (PE). We evaluated the sFlt-1/PlGF ratio as a predictive marker for early-onset PE in women at risk of PE. METHODS This prospective, Spanish, multicenter study included pregnant women with a risk factor for PE, including intrauterine growth restriction, PE, eclampsia or hemolysis, elevated liver enzymes and low platelet count syndrome in previous pregnancy, pregestational diabetes or abnormal uterine artery Doppler. The primary objective was to show that the sFlt-1/PlGF ratio at 20, 24 and 28 weeks' gestation was predictive of early-onset PE (< 34 + 0 weeks). Serum sFlt-1 and PlGF were measured at 20, 24 and 28 weeks. Multivariate logistic regression was used to develop a predictive model. RESULTS A total of 819 women were enrolled, of which 729 were suitable for analysis. Of these, 78 (10.7%) women developed PE (24 early onset and 54 late onset). Median sFlt-1/PlGF ratio at 20, 24 and 28 weeks was 6.3 (interquartile range (IQR), 4.1-9.3), 4.0 (IQR, 2.6-6.3) and 3.3 (IQR, 2.0-5.9), respectively, for women who did not develop PE (controls); 14.5 (IQR, 5.5-43.7), 18.4 (IQR, 8.2-57.9) and 51.9 (IQR, 11.5-145.6) for women with early-onset PE; and 6.7 (IQR, 4.6-9.9), 4.7 (IQR, 2.8-7.2) and 6.0 (IQR, 3.8-10.5) for women with late-onset PE. Compared with early-onset PE, the sFlt-1/PlGF ratio was significantly lower in controls (P < 0.001 at each timepoint) and in women with chronic hypertension (P < 0.001 at each timepoint), gestational hypertension (P < 0.001 at each timepoint) and late-onset PE (P < 0.001 at each timepoint). A prediction model for early-onset PE was developed, which included the sFlt-1/PlGF ratio plus mean arterial pressure, being parous and previous PE, with areas under the receiver-operating characteristics curves of 0.86 (95% CI, 0.77-0.95), 0.91 (95% CI, 0.85-0.97) and 0.93 (95% CI, 0.86-0.99) at 20, 24 and 28 weeks, respectively, and was superior to models using the sFlt-1/PlGF ratio alone or uterine artery mean pulsatility index. CONCLUSIONS The sFlt-1/PlGF ratio can improve prediction of early-onset PE for women at risk of this condition. Copyright © 2017 ISUOG. Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- A. Perales
- Hospital Universitario y Politécnico La FeValenciaSpain
| | | | | | | | | | | | | | - B. Laíz
- Hospital Universitario y Politécnico La FeValenciaSpain
| | - M. Duque
- Hospital Universitario La PazMadridSpain
| | - M. Navarro
- Hospital Universitario Materno Infantil de CanariasGran CanariaSpain
| | - P. Calmarza
- Hospital Universitario Miguel ServetZaragozaSpain
| | - M. Hund
- Roche Diagnostics International LtdRotkreuzSwitzerland
| | - F. V. Álvarez
- Hospital Universitario Central de AsturiasOviedoSpain
| |
Collapse
|
957
|
Saleh L, Vergouwe Y, van den Meiracker AH, Verdonk K, Russcher H, Bremer HA, Versendaal HJ, Steegers EAP, Danser AHJ, Visser W. Angiogenic Markers Predict Pregnancy Complications and Prolongation in Preeclampsia: Continuous Versus Cutoff Values. Hypertension 2017; 70:1025-1033. [PMID: 28847893 DOI: 10.1161/hypertensionaha.117.09913] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/10/2017] [Accepted: 08/09/2017] [Indexed: 02/07/2023]
Abstract
To assess the incremental value of a single determination of the serum levels of sFlt-1 (soluble Fms-like tyrosine kinase 1) and PlGF (placental growth factor) or their ratio, without using cutoff values, for the prediction of maternal and fetal/neonatal complications and pregnancy prolongation, 620 women with suspected/confirmed preeclampsia, aged 18 to 48 years, were included in a prospective, multicenter, observational cohort study. Women had singleton pregnancies and a median pregnancy duration of 34 (range, 20-41) weeks. Complications occurred in 118 women and 248 fetuses. The median duration between admission and delivery was 12 days. To predict prolongation, PlGF showed the highest incremental value (R2=0.72) on top of traditional predictors (gestational age at inclusion, diastolic blood pressure, proteinuria, creatinine, uric acid, alanine transaminase, lactate dehydrogenase, and platelets) compared with R2=0.53 for the traditional predictors only. sFlt-1 showed the highest value to discriminate women with and without maternal complications (C-index=0.83 versus 0.72 for the traditional predictors only), and the sFlt-1/PlGF ratio showed the highest value to discriminate fetal/neonatal complications (C-index=0.86 versus 0.78 for the traditional predictors only). Applying previously suggested cutoff values for the sFlt-1/PlGF ratio yielded lower incremental values than applying continuous values. In conclusion, sFlt-1 and PlGF are strong and independent predictors for days until delivery along with maternal and fetal/neonatal complications on top of the traditional criteria. Their use as continuous variables (instead of applying cutoff values for different gestational ages) should now be tested in a prospective manner, making use of an algorithm calculating the risk of an individual woman with suspected/confirmed preeclampsia to develop complications.
Collapse
Affiliation(s)
- Langeza Saleh
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.S., A.H.v.d.M., A.H.J.D., W.V.), Division Obstetrics and Prenatal Medicine, Department of Obstetrics and Gynecology (L.S., E.A.P.S., W.V.), Department of Public Health, Centre for Medical Decision Sciences (Y.V.), and Department of Clinical Chemistry (H.R.), Erasmus MC, Rotterdam, The Netherlands; Department of Internal Medicine, Ikazia Ziekenhuis, Rotterdam, The Netherlands (K.V.); Department of Obstetrics and Gynecology, Reinier de Graaf Hospital, Delft, The Netherlands (H.A.B.); and Department of Obstetrics and Gynecology, Maasstad Hospital, Rotterdam, The Netherlands (H.J.V.)
| | - Yvonne Vergouwe
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.S., A.H.v.d.M., A.H.J.D., W.V.), Division Obstetrics and Prenatal Medicine, Department of Obstetrics and Gynecology (L.S., E.A.P.S., W.V.), Department of Public Health, Centre for Medical Decision Sciences (Y.V.), and Department of Clinical Chemistry (H.R.), Erasmus MC, Rotterdam, The Netherlands; Department of Internal Medicine, Ikazia Ziekenhuis, Rotterdam, The Netherlands (K.V.); Department of Obstetrics and Gynecology, Reinier de Graaf Hospital, Delft, The Netherlands (H.A.B.); and Department of Obstetrics and Gynecology, Maasstad Hospital, Rotterdam, The Netherlands (H.J.V.)
| | - Anton H van den Meiracker
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.S., A.H.v.d.M., A.H.J.D., W.V.), Division Obstetrics and Prenatal Medicine, Department of Obstetrics and Gynecology (L.S., E.A.P.S., W.V.), Department of Public Health, Centre for Medical Decision Sciences (Y.V.), and Department of Clinical Chemistry (H.R.), Erasmus MC, Rotterdam, The Netherlands; Department of Internal Medicine, Ikazia Ziekenhuis, Rotterdam, The Netherlands (K.V.); Department of Obstetrics and Gynecology, Reinier de Graaf Hospital, Delft, The Netherlands (H.A.B.); and Department of Obstetrics and Gynecology, Maasstad Hospital, Rotterdam, The Netherlands (H.J.V.)
| | - Koen Verdonk
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.S., A.H.v.d.M., A.H.J.D., W.V.), Division Obstetrics and Prenatal Medicine, Department of Obstetrics and Gynecology (L.S., E.A.P.S., W.V.), Department of Public Health, Centre for Medical Decision Sciences (Y.V.), and Department of Clinical Chemistry (H.R.), Erasmus MC, Rotterdam, The Netherlands; Department of Internal Medicine, Ikazia Ziekenhuis, Rotterdam, The Netherlands (K.V.); Department of Obstetrics and Gynecology, Reinier de Graaf Hospital, Delft, The Netherlands (H.A.B.); and Department of Obstetrics and Gynecology, Maasstad Hospital, Rotterdam, The Netherlands (H.J.V.)
| | - Henk Russcher
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.S., A.H.v.d.M., A.H.J.D., W.V.), Division Obstetrics and Prenatal Medicine, Department of Obstetrics and Gynecology (L.S., E.A.P.S., W.V.), Department of Public Health, Centre for Medical Decision Sciences (Y.V.), and Department of Clinical Chemistry (H.R.), Erasmus MC, Rotterdam, The Netherlands; Department of Internal Medicine, Ikazia Ziekenhuis, Rotterdam, The Netherlands (K.V.); Department of Obstetrics and Gynecology, Reinier de Graaf Hospital, Delft, The Netherlands (H.A.B.); and Department of Obstetrics and Gynecology, Maasstad Hospital, Rotterdam, The Netherlands (H.J.V.)
| | - Henk A Bremer
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.S., A.H.v.d.M., A.H.J.D., W.V.), Division Obstetrics and Prenatal Medicine, Department of Obstetrics and Gynecology (L.S., E.A.P.S., W.V.), Department of Public Health, Centre for Medical Decision Sciences (Y.V.), and Department of Clinical Chemistry (H.R.), Erasmus MC, Rotterdam, The Netherlands; Department of Internal Medicine, Ikazia Ziekenhuis, Rotterdam, The Netherlands (K.V.); Department of Obstetrics and Gynecology, Reinier de Graaf Hospital, Delft, The Netherlands (H.A.B.); and Department of Obstetrics and Gynecology, Maasstad Hospital, Rotterdam, The Netherlands (H.J.V.)
| | - Hans J Versendaal
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.S., A.H.v.d.M., A.H.J.D., W.V.), Division Obstetrics and Prenatal Medicine, Department of Obstetrics and Gynecology (L.S., E.A.P.S., W.V.), Department of Public Health, Centre for Medical Decision Sciences (Y.V.), and Department of Clinical Chemistry (H.R.), Erasmus MC, Rotterdam, The Netherlands; Department of Internal Medicine, Ikazia Ziekenhuis, Rotterdam, The Netherlands (K.V.); Department of Obstetrics and Gynecology, Reinier de Graaf Hospital, Delft, The Netherlands (H.A.B.); and Department of Obstetrics and Gynecology, Maasstad Hospital, Rotterdam, The Netherlands (H.J.V.)
| | - Eric A P Steegers
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.S., A.H.v.d.M., A.H.J.D., W.V.), Division Obstetrics and Prenatal Medicine, Department of Obstetrics and Gynecology (L.S., E.A.P.S., W.V.), Department of Public Health, Centre for Medical Decision Sciences (Y.V.), and Department of Clinical Chemistry (H.R.), Erasmus MC, Rotterdam, The Netherlands; Department of Internal Medicine, Ikazia Ziekenhuis, Rotterdam, The Netherlands (K.V.); Department of Obstetrics and Gynecology, Reinier de Graaf Hospital, Delft, The Netherlands (H.A.B.); and Department of Obstetrics and Gynecology, Maasstad Hospital, Rotterdam, The Netherlands (H.J.V.)
| | - A H Jan Danser
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.S., A.H.v.d.M., A.H.J.D., W.V.), Division Obstetrics and Prenatal Medicine, Department of Obstetrics and Gynecology (L.S., E.A.P.S., W.V.), Department of Public Health, Centre for Medical Decision Sciences (Y.V.), and Department of Clinical Chemistry (H.R.), Erasmus MC, Rotterdam, The Netherlands; Department of Internal Medicine, Ikazia Ziekenhuis, Rotterdam, The Netherlands (K.V.); Department of Obstetrics and Gynecology, Reinier de Graaf Hospital, Delft, The Netherlands (H.A.B.); and Department of Obstetrics and Gynecology, Maasstad Hospital, Rotterdam, The Netherlands (H.J.V.).
| | - Willy Visser
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.S., A.H.v.d.M., A.H.J.D., W.V.), Division Obstetrics and Prenatal Medicine, Department of Obstetrics and Gynecology (L.S., E.A.P.S., W.V.), Department of Public Health, Centre for Medical Decision Sciences (Y.V.), and Department of Clinical Chemistry (H.R.), Erasmus MC, Rotterdam, The Netherlands; Department of Internal Medicine, Ikazia Ziekenhuis, Rotterdam, The Netherlands (K.V.); Department of Obstetrics and Gynecology, Reinier de Graaf Hospital, Delft, The Netherlands (H.A.B.); and Department of Obstetrics and Gynecology, Maasstad Hospital, Rotterdam, The Netherlands (H.J.V.)
| |
Collapse
|
958
|
Lucovnik M, Lackner HK, Papousek I, Schmid-Zalaudek K, Schulter G, Roessler A, Moertl MG. Systemic vascular resistance and endogenous inhibitors of nitric oxide synthesis in early- compared to late-onset preeclampsia: preliminary findings. Hypertens Pregnancy 2017; 36:276-281. [DOI: 10.1080/10641955.2017.1364381] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Miha Lucovnik
- Department of Perinatology, Division of Obstetrics and Gynecology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | | | - Ilona Papousek
- Department of Psychology, Biological Psychology Unit, University of Graz, Graz, Austria
| | | | - Guenter Schulter
- Department of Psychology, Biological Psychology Unit, University of Graz, Graz, Austria
| | - Andreas Roessler
- Department of Physiology, Medical University of Graz, Graz, Austria
| | | |
Collapse
|
959
|
Integrative single-cell and cell-free plasma RNA transcriptomics elucidates placental cellular dynamics. Proc Natl Acad Sci U S A 2017; 114:E7786-E7795. [PMID: 28830992 DOI: 10.1073/pnas.1710470114] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The human placenta is a dynamic and heterogeneous organ critical in the establishment of the fetomaternal interface and the maintenance of gestational well-being. It is also the major source of cell-free fetal nucleic acids in the maternal circulation. Placental dysfunction contributes to significant complications, such as preeclampsia, a potentially lethal hypertensive disorder during pregnancy. Previous studies have identified significant changes in the expression profiles of preeclamptic placentas using whole-tissue analysis. Moreover, studies have shown increased levels of targeted RNA transcripts, overall and placental contributions in maternal cell-free nucleic acids during pregnancy progression and gestational complications, but it remains infeasible to noninvasively delineate placental cellular dynamics and dysfunction at the cellular level using maternal cell-free nucleic acid analysis. In this study, we addressed this issue by first dissecting the cellular heterogeneity of the human placenta and defined individual cell-type-specific gene signatures by analyzing more than 24,000 nonmarker selected cells from full-term and early preeclamptic placentas using large-scale microfluidic single-cell transcriptomic technology. Our dataset identified diverse cellular subtypes in the human placenta and enabled reconstruction of the trophoblast differentiation trajectory. Through integrative analysis with maternal plasma cell-free RNA, we resolved the longitudinal cellular dynamics of hematopoietic and placental cells in pregnancy progression. Furthermore, we were able to noninvasively uncover the cellular dysfunction of extravillous trophoblasts in early preeclamptic placentas. Our work showed the potential of integrating transcriptomic information derived from single cells into the interpretation of cell-free plasma RNA, enabling the noninvasive elucidation of cellular dynamics in complex pathological conditions.
Collapse
|
960
|
Fakhouri F, Zuber J, Frémeaux-Bacchi V, Loirat C. Haemolytic uraemic syndrome. Lancet 2017; 390:681-696. [PMID: 28242109 DOI: 10.1016/s0140-6736(17)30062-4] [Citation(s) in RCA: 341] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 10/19/2016] [Accepted: 10/25/2016] [Indexed: 12/17/2022]
Abstract
Haemolytic uraemic syndrome is a form of thrombotic microangiopathy affecting predominantly the kidney and characterised by a triad of thrombocytopenia, mechanical haemolytic anaemia, and acute kidney injury. The term encompasses several disorders: shiga toxin-induced and pneumococcus-induced haemolytic uraemic syndrome, haemolytic uraemic syndrome associated with complement dysregulation or mutation of diacylglycerol kinase ɛ, haemolytic uraemic syndrome related to cobalamin C defect, and haemolytic uraemic syndrome secondary to a heterogeneous group of causes (infections, drugs, cancer, and systemic diseases). In the past two decades, experimental, genetic, and clinical studies have helped to decipher the pathophysiology of these various forms of haemolytic uraemic syndrome and undoubtedly improved diagnostic approaches. Moreover, a specific mechanism-based treatment has been made available for patients affected by atypical haemolytic uraemic syndrome due to complement dysregulation. Such treatment is, however, still absent for several other disease types, including shiga toxin-induced haemolytic uraemic syndrome.
Collapse
Affiliation(s)
- Fadi Fakhouri
- Department of Nephrology, Centre Hospitalier Universitaire, and INSERM UMR S1064, Nantes, France
| | - Julien Zuber
- Assistance Publique-Hôpitaux de Paris, Department of Nephrology and Renal Transplantation, Hôpital Necker, Université Paris Descartes, Paris, France
| | - Véronique Frémeaux-Bacchi
- Assistance Publique-Hôpitaux de Paris, Department of Biological Immunology, Hôpital Européen Georges Pompidou, and INSERM UMR S1138, Complément et Maladies, Centre de Recherche des Cordeliers, Paris, France
| | - Chantal Loirat
- Assistance Publique-Hôpitaux de Paris, Department of Pediatric Nephrology, Hôpital Robert Debré, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
961
|
Hoffmann J, Ossada V, Weber M, Stepan H. An intermediate sFlt-1/PlGF ratio indicates an increased risk for adverse pregnancy outcome. Pregnancy Hypertens 2017; 10:165-170. [PMID: 29153672 DOI: 10.1016/j.preghy.2017.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 08/07/2017] [Accepted: 08/10/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The sFlt-1/PlGF ratio is a valid marker in diagnosing or excluding preeclampsia. The currently used cut offs frame an intermediate zone of 33-85 (<340 weeks) or 33-110 (≥340 weeks), respectively. In this study we sought to evaluate the relevance of an intermediate sFlt-1/PlGF ratio for the clinical pregnancy course and outcome. MATERIAL AND METHODS We retrospectively analysed 533 consecutive patients with sFlt-1/PlGF ratio measurements for suspected preeclampsia. In patients with an intermediate sFlt-1/PlGF ratio, fetal and maternal characteristics and also pregnancy outcome were documented. Furthermore, we compared the patient groups with <340/340-366/≥370 gestational weeks at first visit. RESULTS 83/533 (15.6%) patients had an intermediate sFlt-1/PlGF ratio. Maternal or fetal diseases or twin pregnancies occurred in 87.9%. Preeclampsia/HELLP syndrome developed in 31.3% but were mostly mild or moderate (65.4%). However, severe adverse outcome was observed in 36.1% with severe preeclampsia in 10.8%. Even if further pregnancy duration and gestational week correlated negatively (r=-0.424; p<0.001), 92% of patients, tested with <340 weeks delivered prematurely. The overall preterm birth rate was 27.7%. CONCLUSIONS Patients with an intermediate sFlt-1/PlGF ratio are at risk for severe adverse outcome. An intermediate sFlt-1/PlGF ratio indicates a risk for preterm birth, independent from the occurrence of preeclampsia.
Collapse
Affiliation(s)
- Janine Hoffmann
- Department of Obstetrics, University Hospital Leipzig, Leipzig, Germany.
| | - Victoria Ossada
- Department of Obstetrics, University Hospital Leipzig, Leipzig, Germany
| | - Marie Weber
- Department of Obstetrics, University Hospital Leipzig, Leipzig, Germany
| | - Holger Stepan
- Department of Obstetrics, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
962
|
The influence of prenatal exercise and pre-eclampsia on maternal vascular function. Clin Sci (Lond) 2017; 131:2223-2240. [PMID: 28798074 DOI: 10.1042/cs20171036] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/03/2017] [Accepted: 07/06/2017] [Indexed: 01/10/2023]
Abstract
During healthy pregnancy, the cardiovascular system undergoes diverse adaptations to support adequate transfer of oxygen and nutrients from mother to fetus. In order to accommodate the large expansion of blood volume and associated cardiac output, the structure, mechanics, and function of the arteries are altered. Specifically, in healthy pregnancy there is a remodeling of arteries (increased angiogenesis and vasodilation), a generalized reduction in arterial stiffness (increased compliance), and an enhanced endothelial function. The development of pregnancy complications, specifically pre-eclampsia, is associated with poor placentation (decreased angiogenesis), increased arterial stiffness, and vascular dysfunction (reduced endothelial function). Many of the positive adaptations that occur in healthy pregnancy are enhanced in response to chronic exercise. Specifically, placental angiogenesis and endothelial function have been shown to improve to a greater extent in women who are active during their pregnancy compared with those who are not. Prenatal exercise may be important in helping to reduce the risk of vascular dysfunction in pregnancy. However, our knowledge of the vascular adaptations resulting from maternal exercise is limited. This review highlights maternal vascular adaptations occurring during healthy pregnancy, and contrasts the vascular maladaptation associated with pre-eclampsia. Finally, we discuss the role of prenatal exercise on vascular function in the potential prevention of vascular complications associated with pre-eclampsia.
Collapse
|
963
|
Tsatsaris V. [What place for the biomarkers of preeclampsia?]. ACTA ACUST UNITED AC 2017; 45:385-386. [PMID: 28784350 DOI: 10.1016/j.gofs.2017.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Indexed: 11/25/2022]
Affiliation(s)
- V Tsatsaris
- Université Paris Descartes, CHU Cochin Broca Hôtel-Dieu, Assistance publique-Hôpital de Paris, 3, avenue de l'Observatoire, 75014 Paris, France.
| |
Collapse
|
964
|
Neurological function in children born to preeclamptic and hypertensive mothers - A systematic review. Pregnancy Hypertens 2017; 10:1-6. [PMID: 29153658 DOI: 10.1016/j.preghy.2017.07.144] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/14/2017] [Accepted: 07/21/2017] [Indexed: 11/24/2022]
Abstract
BACKGROUND Offspring whose mothers developed preeclampsia (PE-F1s) show developmental effects that are now being identified, such as cognitive, behavioural, and mood differences compared to offspring from non-complicated pregnancies. We hypothesize that the progressive angiokine dysregulation associated with development of preeclampsia (PE) reflects gene dysregulation in pre-implantation conceptuses, and manifests in all developing fetal tissues rather than exclusively to the placenta. This hypothesis predicts that fetal cerebrovascular and brain development are deviated by fetal-intrinsic, brain-based mechanisms during what is currently considered a placentally-induced maternal disease. Due to our initial results from brain-imaging and cognitive screening in a child pilot PE-F1 cohort, we developed this systematic review to answer the question of whether any consistent neurological measurements have been found to discriminate between brain functions in offspring of mothers who experienced a hypertensive pregnancy vs. offspring of mothers that did not. METHODS Relevant studies were searched systematically up to June 2017 in MEDLINE, PsycINFO, EMBASE and the grey literature. RESULTS Following predetermined inclusion and exclusion criteria, our search identified 27 out of 464 studies reporting on neurological function in offspring born to preeclamptic and hypertensive mothers. CONCLUSION The current literature strongly supports the conclusion of the behavioural and cognitive deviations in PE-F1s. However, only three studies associated their findings with brain measurements via magnetic resonance imaging (MRI) in both healthy and at-risk pediatric populations. PE-F1s should be identified as an at-risk pediatric population during brain development and studied further as a defined group, perhaps stratified by maternal plasma angiokine levels.
Collapse
|
965
|
Shinohara S, Uchida Y, Kasai M, Sunami R. Association between the high soluble fms-like tyrosine kinase-1 to placental growth factor ratio and adverse outcomes in asymptomatic women with early-onset fetal growth restriction. Hypertens Pregnancy 2017; 36:269-275. [DOI: 10.1080/10641955.2017.1334800] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Satoshi Shinohara
- Department of Obstetrics and Gynecology, Yamanashi Prefectural Central Hospital, Kofu, Yamanashi, Fujimi, Japan
| | - Yuzo Uchida
- Department of Obstetrics and Gynecology, Yamanashi Prefectural Central Hospital, Kofu, Yamanashi, Fujimi, Japan
| | - Mayuko Kasai
- Department of Obstetrics and Gynecology, Yamanashi Prefectural Central Hospital, Kofu, Yamanashi, Fujimi, Japan
| | - Rei Sunami
- Department of Obstetrics and Gynecology, Yamanashi Prefectural Central Hospital, Kofu, Yamanashi, Fujimi, Japan
| |
Collapse
|
966
|
Endothelial Dysfunction in Severe Preeclampsia is Mediated by Soluble Factors, Rather than Extracellular Vesicles. Sci Rep 2017; 7:5887. [PMID: 28725005 PMCID: PMC5517616 DOI: 10.1038/s41598-017-06178-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 06/09/2017] [Indexed: 11/24/2022] Open
Abstract
In severe early-onset preeclampsia (sPE) the placenta releases soluble angiogenesis-regulating proteins, trophoblast-derived fragments, and extracellular vesicles (EVs). Their relative importance in disease pathogenesis is not presently understood. We explanted placental villi from healthy and sPE women then separated the media into: total-conditioned, EV-depleted and EV-enriched media. Three fractions were compared for; angiogenic protein secretion by ELISA, angiogenic and inflammation gene mRNA expression and leukocyte adhesion assay. sPE placental villi secreted significantly less PlGF (70 ± 18 pg/mL) than preterm controls (338 ± 203; p = 0.03). sFlt-1:PlGF ratios in total-conditioned (115 ± 29) and EV-depleted media (136 ± 40) from sPE placental villi were significantly higher than in EV-enriched media (42 ± 12; p < 0.01) or any preterm or term media. Fluorescent-labeled EVs derived across normal gestation, but not from sPE, actively entered HUVECs. From sPE placental villi, the soluble fraction, but not EV-enriched fraction, significantly repressed angiogenesis (0.83 ± 0.05 fold, p = 0.02), induced HO-1 mRNA (15.3 ± 5.1 fold, p < 0.05) and induced leukocyte adhesion (2.2 ± 0.4 fold, p = 0.04). Soluble media (total-conditioned and EV-depleted media) from sPE placental villi induced endothelial dysfunction in HUVEC, while the corresponding EV-enriched fraction showed no such effects. Our data suggest that soluble factors including angiogenesis-regulating proteins, dominate the vascular pathology of this disease.
Collapse
|
967
|
Introduction: Women's Renal Health Across the Decades. Semin Nephrol 2017; 37:309-310. [PMID: 28711068 DOI: 10.1016/j.semnephrol.2017.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
968
|
Saleh L, Samantar R, Garrelds IM, van den Meiracker AH, Visser W, Danser AHJ. Low Soluble Fms-Like Tyrosine Kinase-1, Endoglin, and Endothelin-1 Levels in Women With Confirmed or Suspected Preeclampsia Using Proton Pump Inhibitors. Hypertension 2017; 70:594-600. [PMID: 28716993 DOI: 10.1161/hypertensionaha.117.09741] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 05/31/2017] [Accepted: 06/14/2017] [Indexed: 01/01/2023]
Abstract
Patients with preeclampsia display elevated placenta-derived sFlt-1 (soluble Fms-like tyrosine kinase-1) and endoglin levels and decreased placental growth factor levels. Proton pump inhibitors (PPIs) decrease trophoblast sFlt-1 and endoglin secretion in vitro. PPIs are used during pregnancy to combat reflux disease. Here, we investigated whether PPIs affect sFlt-1 in women with confirmed/suspected preeclampsia, making use of a prospective cohort study involving 430 women. Of these women, 40 took PPIs (6 esomeprazole, 32 omeprazole, and 2 pantoprazole) for 8 to 45 (median 29) days before sFlt-1 measurement. Measurements were only made once, at study entry between weeks 20 and 41 (median 33 weeks). PPI use was associated with lower sFlt-1 levels, with no change in placental growth factor levels, both when compared with all non-PPI users and with 80 gestational age-matched controls selected from the non-PPI users. No sFlt-1/placental growth factor alterations were observed in women using ferrous fumarate or macrogol while, as expected, women using antihypertensive medication displayed higher sFlt-1 levels and lower placental growth factor levels. The PPI use-associated decrease in sFlt-1 was independent of the application of antihypertensive drugs and also occurred when restricting our analysis to patients with hypertensive disease of pregnancy at study entry. PPI users displayed more cases with preexisting proteinuria, less gestational hypertension, and a lower number of neonatal sepsis cases. Finally, their plasma endoglin and endothelin-1 levels were lower while sFlt-1 levels correlated positively with both. In conclusion, PPI use associates with low sFlt-1, endoglin, and endothelin-1 levels, warranting prospective trials to investigate the therapeutic potential of PPIs in preeclampsia.
Collapse
Affiliation(s)
- Langeza Saleh
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.S., I.M.G., A.H.v.d.M., W.V., A.H.J.D.) and Division Obstetrics and Prenatal Medicine, Department of Obstetrics and Gynaecology (L.S., R.S., W.V.), Erasmus MC, Rotterdam, The Netherlands
| | - Raaho Samantar
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.S., I.M.G., A.H.v.d.M., W.V., A.H.J.D.) and Division Obstetrics and Prenatal Medicine, Department of Obstetrics and Gynaecology (L.S., R.S., W.V.), Erasmus MC, Rotterdam, The Netherlands
| | - Ingrid M Garrelds
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.S., I.M.G., A.H.v.d.M., W.V., A.H.J.D.) and Division Obstetrics and Prenatal Medicine, Department of Obstetrics and Gynaecology (L.S., R.S., W.V.), Erasmus MC, Rotterdam, The Netherlands
| | - Anton H van den Meiracker
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.S., I.M.G., A.H.v.d.M., W.V., A.H.J.D.) and Division Obstetrics and Prenatal Medicine, Department of Obstetrics and Gynaecology (L.S., R.S., W.V.), Erasmus MC, Rotterdam, The Netherlands
| | - Willy Visser
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.S., I.M.G., A.H.v.d.M., W.V., A.H.J.D.) and Division Obstetrics and Prenatal Medicine, Department of Obstetrics and Gynaecology (L.S., R.S., W.V.), Erasmus MC, Rotterdam, The Netherlands
| | - A H Jan Danser
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.S., I.M.G., A.H.v.d.M., W.V., A.H.J.D.) and Division Obstetrics and Prenatal Medicine, Department of Obstetrics and Gynaecology (L.S., R.S., W.V.), Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
969
|
Jin X, Xu Z, Cao J, Shao P, Zhou M, Qin Z, Liu Y, Yu F, Zhou X, Ji W, Cai W, Ma Y, Wang C, Shan N, Yang N, Chen X, Li Y. Proteomics analysis of human placenta reveals glutathione metabolism dysfunction as the underlying pathogenesis for preeclampsia. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017; 1865:1207-1214. [PMID: 28705740 DOI: 10.1016/j.bbapap.2017.07.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 07/03/2017] [Accepted: 07/06/2017] [Indexed: 12/19/2022]
Abstract
Hypertensive disorder in pregnancy (HDP) refers to a series of diseases that cause the hypertension during pregnancy, including HDP, preeclampsia (PE) and eclampsia. This study screens differentially expressed proteins of placenta tissues in PE cases using 2D LC-MS/MS quantitative proteomics strategy. A total of 2281 proteins are quantified, of these, 145 altering expression proteins are successfully screened between PE and control cases (p<0.05). Bioinformatics analysis suggests that these proteins are mainly involved in many biological processes, such as oxidation reduction, mitochondrion organization, and acute inflammatory response. Especially, the glutamine metabolic process related molecules, GPX1, GPX3, SMS, GGCT, GSTK1, NFκB, GSTT2, SOD1 and GCLM, are involved in the switching process from oxidized glutathione (GSSG) conversion to the reduced glutathione (GSH) by glutathione, mercapturic acid and arginine metabolism process. Results of this study revealed that glutathione metabolism disorder of placenta tissues may contribute to the occurrence of PE disease.
Collapse
Affiliation(s)
- Xiaohan Jin
- Tianjin Key Laboratory of Cardiovascular Remodeling & Target Organ Injury, Pingjin Hospital Heart Center, Tianjin 300162, China; Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, China
| | - Zhongwei Xu
- Tianjin Key Laboratory of Cardiovascular Remodeling & Target Organ Injury, Pingjin Hospital Heart Center, Tianjin 300162, China; Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, China
| | - Jin Cao
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, China
| | - Ping Shao
- Women and Children Health Care Center, Tianjin 300070, China
| | - Maobin Zhou
- Tianjin Key Laboratory of Cardiovascular Remodeling & Target Organ Injury, Pingjin Hospital Heart Center, Tianjin 300162, China
| | - Zhe Qin
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, China
| | - Yan Liu
- Tianjin First Center Hospital, Tianjin 300192, China
| | - Fang Yu
- Obstetrics and Gynecology Department, Pingjin Hospital, Tianjin 300162, China
| | - Xin Zhou
- Tianjin Key Laboratory of Cardiovascular Remodeling & Target Organ Injury, Pingjin Hospital Heart Center, Tianjin 300162, China
| | - Wenjie Ji
- Tianjin Key Laboratory of Cardiovascular Remodeling & Target Organ Injury, Pingjin Hospital Heart Center, Tianjin 300162, China
| | - Wei Cai
- Tianjin Key Laboratory of Cardiovascular Remodeling & Target Organ Injury, Pingjin Hospital Heart Center, Tianjin 300162, China
| | - Yongqiang Ma
- Tianjin Key Laboratory of Cardiovascular Remodeling & Target Organ Injury, Pingjin Hospital Heart Center, Tianjin 300162, China
| | - Chengyan Wang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, China
| | - Nana Shan
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, China
| | - Ning Yang
- Tianjin Key Laboratory of Cardiovascular Remodeling & Target Organ Injury, Pingjin Hospital Heart Center, Tianjin 300162, China
| | - Xu Chen
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin 300100, China.
| | - Yuming Li
- Tianjin Key Laboratory of Cardiovascular Remodeling & Target Organ Injury, Pingjin Hospital Heart Center, Tianjin 300162, China.
| |
Collapse
|
970
|
Vigneron C, Hertig A. Micro-angiopathies thrombotiques du péripartum : physiopathologie, diagnostic et traitement. MEDECINE INTENSIVE REANIMATION 2017. [DOI: 10.1007/s13546-017-1287-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
971
|
Park S, Lee SM, Park JS, Hong JS, Chin HJ, Na KY, Kim DK, Oh KH, Joo KW, Kim YS, Lee H. Midterm eGFR and Adverse Pregnancy Outcomes: The Clinical Significance of Gestational Hyperfiltration. Clin J Am Soc Nephrol 2017; 12:1048-1056. [PMID: 28611078 PMCID: PMC5498359 DOI: 10.2215/cjn.12101116] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 03/23/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND OBJECTIVES Although hemodynamic adaptation plays a crucial role in maintaining gestation, the clinical significance of midterm renal hyperfiltration (MRH) on pregnancy outcomes is unknown. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS This was an observational cohort study. Women with a singleton pregnancy and a serum creatinine measurement during their second trimester were followed at two university hospitals in Korea between 2001 and 2015. Those with substantial renal function impairment or who delivered during the second trimester were not considered. MRH was represented by the highest eGFR, which was calculated using the Chronic Kidney Disease Epidemiology Collaboration method. An adverse pregnancy event was defined by the composition of preterm birth (gestational age <37 weeks), low birth weight (<2.5 kg), and preeclampsia. RESULTS Data from 1931 pregnancies were included. The relationship between midterm eGFR and adverse pregnancy outcomes, which occurred in 538 mothers, was defined by a nonlinear U-shaped curve. The adjusted odds ratio and associated 95% confidence interval (95% CI) of an adverse pregnancy outcome for eGFR levels below and above the reference level of 120-150 ml/min per 1.73 m2 were 1.97 (95% CI, 1.34 to 2.89; P<0.001) for ≥150 ml/min per 1.73 m2; 1.57 (95% CI, 1.23 to 2.00; P<0.001) for 90-120 ml/min per 1.73 m2; and 4.93 (95% CI, 1.97 to 12.31; P<0.001) for 60-90 ml/min per 1.73 m2. Moreover, among mothers without baseline CKD, women with adverse pregnancy outcomes had less prominent MRH than those without (P<0.001). CONCLUSIONS We identified a unique U-shaped relationship between midterm eGFR and adverse pregnancy outcomes, and the optimal range of midterm eGFR levels was 120-150 ml/min per 1.73 m2. In those without evident functional renal impairment, the absence of prominent MRH might be a significant risk factor for poor pregnancy outcomes.
Collapse
Affiliation(s)
- Sehoon Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Mi Lee
- Department of Obstetrics and Gynecology, Seoul National College of Medicine, Seoul, Korea
| | - Joong Shin Park
- Department of Obstetrics and Gynecology, Seoul National College of Medicine, Seoul, Korea
| | | | - Ho Jun Chin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea College of Medicine, Seoul, Korea; and
| | - Ki Young Na
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea College of Medicine, Seoul, Korea; and
| | - Dong Ki Kim
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea College of Medicine, Seoul, Korea; and
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kwon Wook Joo
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea College of Medicine, Seoul, Korea; and
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Yon Su Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea College of Medicine, Seoul, Korea; and
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hajeong Lee
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea College of Medicine, Seoul, Korea; and
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
972
|
Wu WB, Xu YY, Cheng WW, Yuan B, Zhao JR, Wang YL, Zhang HJ. Decreased PGF may contribute to trophoblast dysfunction in fetal growth restriction. Reproduction 2017; 154:319-329. [PMID: 28676532 DOI: 10.1530/rep-17-0253] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 06/16/2017] [Accepted: 07/04/2017] [Indexed: 12/12/2022]
Abstract
Fetal growth restriction (FGR) threatens perinatal health and is correlated with increased incidence of fetal original adult diseases. Most cases of FGR were idiopathic, which were supposed to be associated with placental abnormality. Decreased circulating placental growth factor (PGF) was recognized as an indication of placental deficiency in FGR. In this study, the epigenetic regulation of PGF in FGR placentas and the involvement of PGF in modulation of trophoblast activity were investigated. The expression level of PGF in placental tissues was determined by RT-qPCR, immunohistochemistry and ELISA. DNA methylation profile of PGF gene was analyzed by bisulfite sequencing. Trophoblastic cell lines were treated with ZM-306416, an inhibitor of PGF receptor FLT1, to observe the effect of PGF/FLT1 signaling on cell proliferation and migration. We demonstrated that PGF was downregulated in placentas from FGR pregnancies compared with normal controls. The villous expression of PGF was positively correlated with placental and fetal weight. The CpG island inside PGF promoter was hypomethylated without obvious difference in both normal and FGR placentas. However, the higher DNA methylation at another CpG island downstream exon 7 of PGF was demonstrated in FGR placentas. Additionally, we found FLT1 was expressed in trophoblast cells. Inhibition of PGF/FLT1 signaling by a selective inhibitor impaired trophoblast proliferation and migration. In conclusion, our data suggested that the PGF expression was dysregulated, and disrupted PGF/FLT1 signaling in trophoblast might contribute to placenta dysfunction in FGR. Thus, our results support the significant role of PGF in the pathogenesis of FGR.
Collapse
Affiliation(s)
- Wei-Bin Wu
- Departments of Pathology and Bio-Bank, The International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Embryo-Fetal Original Adult Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue-Ying Xu
- Departments of Pathology and Bio-Bank, The International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Wei Cheng
- Department of Obstetrics, The International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Yuan
- Department of Computer Science and Engineer, Shanghai Jiao Tong University, Shanghai, China
| | - Jiu-Ru Zhao
- Departments of Pathology and Bio-Bank, The International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Embryo-Fetal Original Adult Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan-Lin Wang
- Prenatal Diagnosis Center & Fetal Medicine Unit, The International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui-Juan Zhang
- Departments of Pathology and Bio-Bank, The International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Embryo-Fetal Original Adult Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
973
|
Blood pressure in adolescent patients with pre-eclampsia and eclampsia. Int J Gynaecol Obstet 2017; 138:335-339. [DOI: 10.1002/ijgo.12237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/24/2017] [Accepted: 06/07/2017] [Indexed: 11/07/2022]
|
974
|
Is Canada Ready to Adopt Maternal Placental Growth Factor Testing to Improve Clinical Outcomes for Women with Suspected Preeclampsia? JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2017; 39:580-583. [DOI: 10.1016/j.jogc.2016.10.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 10/18/2016] [Accepted: 10/18/2016] [Indexed: 11/19/2022]
|
975
|
Muñoz-Hernández R, Medrano-Campillo P, Miranda ML, Macher HC, Praena-Fernández JM, Vallejo-Vaz AJ, Dominguez-Simeon MJ, Moreno-Luna R, Stiefel P. Total and Fetal Circulating Cell-Free DNA, Angiogenic, and Antiangiogenic Factors in Preeclampsia and HELLP Syndrome. Am J Hypertens 2017; 30:673-682. [PMID: 28338787 DOI: 10.1093/ajh/hpx024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 01/30/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Preeclampsia (PE) is a hypertensive disorder of pregnancy characterized by hypertension and proteinuria. The HELLP syndrome is the most severe form of PE. The aim of the present study was to determine different potential biomarkers that may help us perform an early diagnosis of the disease, assess on the severity of the disease, and/or predict maternal or fetal adverse outcomes. METHODS We measured serum levels of total and fetal circulating cell-free DNA (cfDNA), soluble endoglin, soluble form of vascular endothelial growth factor receptor, and placental growth factor in a healthy control group of pregnant women (n = 26), patients with mild (n = 37) and severe PE (n = 25), and patients with HELLP syndrome (n = 16). RESULTS We observed a gradual and strong relationship between all the biomarkers mentioned and the range of severity of PE, with the highest levels in patients with HELLP syndrome. Nevertheless, only the values of total cfDNA were able to significantly differentiate severe PE and HELLP syndrome (20957 ± 2784 vs. 43184 ± 8647 GE/ml, P = 0.01). Receiver operating characteristic (ROC) curves were constructed (i) for the healthy group with respect to the groups with PE and (ii) for patients with PE with respect to the group with HELLP syndrome; sensitivity and specificity values at different cutoff levels were calculated in each case. The maximum ROC area under the curve value for PE and HELLP syndrome (with respect to controls) was 0.91 (P < 0.001). CONCLUSIONS The measured biomarkers of cell damage, angiogenesis, and antiangiogenesis may reflect the severity of PE, with higher levels in patients who develop HELLP syndrome. In addition, these biomarkers may also help predict adverse fetal and maternal outcomes.
Collapse
Affiliation(s)
- Rocío Muñoz-Hernández
- Laboratorio de Hipertensión Arterial e Hipercolesterolemia, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Laboratorio de Investigación clínica y traslacional en enfermedades hepáticas y digestivas. Instituto de Biomedicina de Sevilla (IBiS). Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Pablo Medrano-Campillo
- Laboratorio de Hipertensión Arterial e Hipercolesterolemia, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Universidad Autónoma de Chile, Chile
| | - Maria L Miranda
- Laboratorio de Hipertensión Arterial e Hipercolesterolemia, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Unidad Clínico-Experimental de Riesgo Vascular (UCERV-UCAMI), Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Hada C Macher
- Servicio de Bioquímica Clínica, Hospital Virgen del Rocío, Sevilla, Spain
| | - Jose Manuel Praena-Fernández
- Unidad de Asesoría Estadística, Metodología y Evaluación de Investigación, Fundación Pública Andaluza para la Gestión de la Investigación en Salud de Sevilla (FISEVI). Sevilla, Spain
| | - Antonio J Vallejo-Vaz
- Laboratorio de Hipertensión Arterial e Hipercolesterolemia, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- School of Public Health, Imperial College London. London, UK
| | - María J Dominguez-Simeon
- Laboratorio de Hipertensión Arterial e Hipercolesterolemia, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Rafael Moreno-Luna
- Laboratorio de Fisiopatología Vascular, Hospital de parapléjicos de Toledo. Toledo, Spain
| | - Pablo Stiefel
- Laboratorio de Hipertensión Arterial e Hipercolesterolemia, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Unidad Clínico-Experimental de Riesgo Vascular (UCERV-UCAMI), Hospital Universitario Virgen del Rocío, Sevilla, Spain
| |
Collapse
|
976
|
Visceral adipose tissue activated macrophage content and inflammatory adipokine secretion is higher in pre-eclampsia than in healthy pregnancys. Clin Sci (Lond) 2017; 131:1529-1540. [PMID: 28566469 PMCID: PMC6376613 DOI: 10.1042/cs20160832] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 05/30/2017] [Accepted: 05/31/2017] [Indexed: 12/11/2022]
Abstract
Obesity increases pre-eclampsia (PE) risk. Adipose tissue inflammation may contribute to the clinical syndrome of PE. We compared adipose tissue macrophage infiltration and release of pro-inflammatory adipokines in PE and healthy pregnancy. Subcutaneous and visceral adipose tissue biopsies were collected from healthy (n=13) and PE (n=13) mothers. Basal and lipopolysaccharide (LPS) stimulated adipocyte TNFα, IL-6, CCL-2, and CRP release was measured. Adipose tissue cell densities of activated (cfms+) and total (CD68+) macrophages were determined. In PE only, visceral adipose tissue TNFα release was increased after LPS stimulation (57 [76] versus 81 [97] pg/ml/µg DNA, P=0.030). Basal TNFα release was negatively correlated insulin sensitivity of visceral adipocytes (r = −0.61, P=0.030) in PE. Visceral adipocyte IL-6 release was increased after LPS stimulation in PE only (566 [696] versus 852 [914] pg/ml/µg DNA, P=0.019). Visceral adipocyte CCL-2 basal (67 [61] versus 187 [219] pg/ml/µgDNA, P=0.049) and stimulated (46 [46] versus 224 [271] pg/ml/µg DNA, P=0.003) release was greater than in subcutaneous adipocytes in PE only. In PE, median TNF mRNA expression in visceral adipose tissue was higher than controls (1.94 [1.13–4.14] versus 0.8 [0.00–1.27] TNF/PPIA ratio, P=0.006). In visceral adipose tissue, CSF1R (a marker of activated macrophages) mRNA expression (24.8[11.0] versus 51.0[29.9] CSF1R/PPIA ratio, P=0.011) and activated (cfms+) macrophage count (6.7[2.6] versus 15.2[8.8] % cfms+/adipocyte, P=0.031) were higher in PE than in controls. In conclusion, our study demonstrates dysregulation of inflammatory pathways predominantly in visceral adipose tissue in PE. Inflammation of visceral adipose tissue may mediate many of the adverse metabolic effects associated with PE.
Collapse
|
977
|
Moe K, Heidecke H, Dechend R, Staff AC. Dysregulation of circulating autoantibodies against VEGF-A, VEGFR-1 and PlGF in preeclampsia - A role in placental and vascular health? Pregnancy Hypertens 2017; 10:83-89. [PMID: 29153696 DOI: 10.1016/j.preghy.2017.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/31/2017] [Accepted: 06/04/2017] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Preeclampsia is a state of antiangiogenesis, with high levels of maternal circulating sVEGFR-1 (soluble vascular endothelial growth factor receptor 1, also named sFlt1) and low levels of PlGF (placenta growth factor). Various autoantibodies have been detected in preeclamptic patients. We hypothesize that circulating autoantibodies against VEGF-A (AA-VEGF-A), VEGFR-1 (AA-VEGFR-1) and PlGF (AA-PlGF) are present in preeclamptic women, with different levels from pregnant women with normotensive pregnancies. Secondly, we wanted to analyze if autoantibody levels are associated to sFlt1 or PLGF levels. STUDY DESIGN Retrospective cross sectional study of 88 women with singleton pregnancies who delivered at Oslo University Hospital of whom 46 had preeclampsia and 42 had uncomplicated normotensive pregnancies. Novel immunoassays for IgG-autoantibodies against VEGFA, VEGFR-1 and PlGF were developed and serum samples were assayed. MAIN OUTCOME MEASURES AND RESULTS AA-VEGF-A, AA-VEGF-R1 and AA-PlGF were significantly lower in preeclamptic pregnancies (n=42) compared to normotensive pregnancies (n=46) (p<0.05). On unadjusted analysis, only AA-VEGFA and AA-VEGFR-1 were predictors of PE, but none were independent predictors after adjusting for BMI (body mass index) and parity. In the subgroup of normotensive and PE women with overlapping sVEGFR-1/PlGF-ratios, AA-VEGF was a significant predictor of PE with AUC: 0.735. CONCLUSION IgG autoantibodies against VEGF-A VEGFR-1 and PlGF can be found in pregnant women. They are dysregulated in preeclampsia. The roles of these autoantibodies are unknown, but this study suggests they play a protective role in pregnancy. The levels of AA against VEGF-A, VEGFR-1 and PlGF might be important factors contributing to anti-angiogenesis regulation.
Collapse
Affiliation(s)
- Kjartan Moe
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway, PB 1171, Blindern, 0381 Oslo, Norway.
| | - Harald Heidecke
- CellTrend GmbH, Im Biotechnologiepark, 14943 Luckenwalde, Germany.
| | - Ralf Dechend
- HELIOS Clinic, Berlin, Germany; Experimental and Clinical Research Center, Charité Medical Faculty and Max-Delbrueck Center for Molecular Medicine, Berlin, Germany.
| | - Anne Cathrine Staff
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway, PB 1171, Blindern, 0381 Oslo, Norway; Department of Obstetrics and Gynaecology, Oslo University Hospital, PB 4956 Nydalen, 0424 Oslo, Norway.
| |
Collapse
|
978
|
Lokki AI, Daly E, Triebwasser M, Kurki MI, Roberson EDO, Häppölä P, Auro K, Perola M, Heinonen S, Kajantie E, Kere J, Kivinen K, Pouta A, Salmon JE, Meri S, Daly M, Atkinson JP, Laivuori H. Protective Low-Frequency Variants for Preeclampsia in the Fms Related Tyrosine Kinase 1 Gene in the Finnish Population. Hypertension 2017; 70:365-371. [PMID: 28652462 DOI: 10.1161/hypertensionaha.117.09406] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 04/04/2017] [Accepted: 05/28/2017] [Indexed: 12/11/2022]
Abstract
Preeclampsia is a common pregnancy-specific vascular disorder characterized by new-onset hypertension and proteinuria during the second half of pregnancy. Predisposition to preeclampsia is in part heritable. It is associated with an increased risk of cardiovascular disease later in life. We have sequenced 124 candidate genes implicated in preeclampsia to pinpoint genetic variants contributing to predisposition to or protection from preeclampsia. First, targeted exomic sequencing was performed in 500 preeclamptic women and 190 controls from the FINNPEC cohort (Finnish Genetics of Preeclampsia Consortium). Then 122 women with a history of preeclampsia and 1905 parous women with no such history from the National FINRISK Study (a large Finnish population survey on risk factors of chronic, noncommunicable diseases) were included in the analyses. We tested 146 rare and low-frequency variants and found an excess (observed 13 versus expected 7.3) nominally associated with preeclampsia (P<0.05). The most significantly associated sequence variants were protective variants rs35832528 (E982A; P=2.49E-4; odds ratio=0.387) and rs141440705 (R54S; P=0.003; odds ratio=0.442) in Fms related tyrosine kinase 1. These variants are enriched in the Finnish population with minor allele frequencies 0.026 and 0.017, respectively. They may also be associated with a lower risk of heart failure in 11 257 FINRISK women. This study provides the first evidence of maternal protective genetic variants in preeclampsia.
Collapse
Affiliation(s)
- A Inkeri Lokki
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.).
| | - Emma Daly
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.)
| | - Michael Triebwasser
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.)
| | - Mitja I Kurki
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.)
| | - Elisha D O Roberson
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.)
| | - Paavo Häppölä
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.)
| | - Kirsi Auro
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.)
| | - Markus Perola
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.)
| | - Seppo Heinonen
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.)
| | - Eero Kajantie
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.)
| | - Juha Kere
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.)
| | - Katja Kivinen
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.)
| | - Anneli Pouta
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.)
| | - Jane E Salmon
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.)
| | - Seppo Meri
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.)
| | - Mark Daly
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.)
| | - John P Atkinson
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.)
| | - Hannele Laivuori
- From the Immunobiology, Research Programs Unit (A.I.L., S.M.), Molecular Neurology, Research Programs Unit (J.K.), and Institute for Molecular Medicine Finland/HiLIFE Unit (P.H., K.A., M.P., H.L.), University of Helsinki, Finland; Medical and Clinical Genetics (A.I.L., H.L.), Bacteriology and Immunology (A.I.L., S.M.), Obstetrics and Gynaecology (K.A., S.H., H.L.), and Children's Hospital (E.K), University of Helsinki and Helsinki University Hospital, Finland; Folkhälsan Institute of Genetics (J.K.), University of Helsinki, Finland; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA (E.D., M.I.K., M.D.); Department of Medicine, Division of Rheumatology (M.T., E.D.O.R., J.P.A.) and Department of Genetics (E.D.O.R.), Washington University School of Medicine, St. Louis, MO; Neurosurgery of Neuro Center, Kuopio University Hospital, Finland (M.I.K.); Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston (M.I.K.); Unit of Genetics and Biomarkers (K.A.), Department of Health (M.P., E.K.), Chronic Disease Prevention Unit, Department of Health (E.K.), and Department of Government Services (A.P.), National Institute for Health and Welfare, Helsinki, Finland; The Estonian Genome Center, University of Tartu, Estonia (M.P.); PEDEGO Research Unit, MRC Oulu, University of Oulu and Oulu University Hospital, Finland (E.K., A.P.); Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden (J.K.); Department of Medical and Molecular Genetics, King's College, London, United Kingdom (J.K.); Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (K.K.); Department of Medicine, Hospital for Special Surgery-Weill Cornell Medicine, New York, NY (J.E.S.); and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston (M.D.).
| |
Collapse
|
979
|
Dai X, Song X, Rui C, Meng L, Xue X, Ding H, Shen R, Li J, Li J, Lu Y, Long W. Peptidome Analysis of Human Serum From Normal and Preeclamptic Pregnancies. J Cell Biochem 2017; 118:4341-4348. [PMID: 28430386 DOI: 10.1002/jcb.26087] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 04/20/2017] [Indexed: 12/14/2022]
Abstract
Preeclampsia is a kind of disease that severely harms the health of pregnant women and infants. To better understand the molecular mechanisms involved in preeclampsia, we used liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS) to construct a comparative peptidomic profiling of human serum between normal and preeclamptic pregnancies. A total of 201 peptides were confidently identified, with 21 up-regulated and three down-regulated. Further analysis indicated that these differentially expressed peptides correlate with enzyme regulator activity, biological regulation, and coagulation cascades occurring during pathological changes of preeclampsia. The identification of key peptides in serum may serve not only as a basis for better understanding and further exploring the etiology and pathogenesis of PE, but also as potential biomarkers and in providing targets for future therapy in PE, especially in early onset severe PE (sPE). J. Cell. Biochem. 118: 4341-4348, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xiaonan Dai
- Department of Obstetrics, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Xuejing Song
- Department of Obstetrics, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China.,Fourth Clinical Medicine College, Nanjing Medical University, Nanjing, China
| | - Can Rui
- Department of Obstetrics, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Li Meng
- Nanjing Maternity and Child Health Medical Institute, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Xuan Xue
- Department of Obstetrics, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Hongjuan Ding
- Department of Obstetrics, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Rong Shen
- Nanjing Maternity and Child Health Medical Institute, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Jun Li
- State Key Laboratory of Reproductive Medicine, Department of Plastic and Cosmetic Surgery, Maternal and Child Health Medical Institute, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Jingyun Li
- State Key Laboratory of Reproductive Medicine, Department of Plastic and Cosmetic Surgery, Maternal and Child Health Medical Institute, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Yuanqing Lu
- Department of Obstetrics, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Wei Long
- Department of Obstetrics, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| |
Collapse
|
980
|
Verlohren S, Perschel FH, Thilaganathan B, Dröge LA, Henrich W, Busjahn A, Khalil A. Angiogenic Markers and Cardiovascular Indices in the Prediction of Hypertensive Disorders of Pregnancy. Hypertension 2017; 69:1192-1197. [DOI: 10.1161/hypertensionaha.117.09256] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/09/2017] [Accepted: 04/06/2017] [Indexed: 01/21/2023]
Abstract
Angiogenic and antiangiogenic factors have proven to be an accurate predictive means of preeclampsia. Echocardiographic studies have shown that women with preeclampsia exhibit significant cardiovascular strain, especially early-onset preeclampsia. The aim of this study is to determine preeclampsia risk with soluble fms-like tyrosin kinase 1/placental growth factor ratio, serum NT-proBNP (N-terminal pro B-type natriuretic peptide), and biophysical markers of cardiovascular function in a prospective case–control study. We examined a cohort of 110 pregnant women with uneventful pregnancy outcome (controls) and 129 with hypertensive pregnancy disorders, including 77 with preeclampsia and 52 with pregnancy-induced hypertension. Cardiac indices were obtained with a USCOM-1A monitor, and soluble fms-like tyrosin kinase 1, placental growth factor, and NT-proBNP were measured in serum samples on automated platforms. Logistic regression, as well as Cox proportional hazard analysis, was performed. There were significant contributions from all variables tested, except for heart rate, stroke volume index, and cardiac index to the prediction model. When testing accuracy of respective markers in combination (full model) versus individual markers (soluble fms-like tyrosin kinase 1/placental growth factor ratio and total peripheral resistance) was compared. The soluble fms-like tyrosin kinase 1/placental growth factor ratio and total peripheral resistance performed as good as the full model, except for hypertensive pregnancy disorders and pregnancy-induced hypertension, where the full model performed better. The additional assessment of biophysical and biochemical markers of cardiovascular strain in pregnancy increases the detection of the composite group of hypertensive pregnancy disorders, while not significantly improving detection of preeclampsia alone. This offers a more precise insight into the pathogenesis of the disease, as well as offering a window for intervention, possibly decreasing cardiovascular mortality in these women.
Collapse
Affiliation(s)
- Stefan Verlohren
- From the Departments of Obstetrics (S.V., L.A.D., W.H.) and Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry (F.H.P.), Charité Universitätsmedizin Berlin, Germany; Labor Berlin - Charité Vivantes GmbH, Germany (F.H.P.); Fetal Medicine Unit, St Georges University Hospital Foundation NHS Trust and St Georges University London, UK (B.T., A.K.); and HealthTwiSt GmbH, Berlin, Germany (A.B.)
| | - Frank H. Perschel
- From the Departments of Obstetrics (S.V., L.A.D., W.H.) and Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry (F.H.P.), Charité Universitätsmedizin Berlin, Germany; Labor Berlin - Charité Vivantes GmbH, Germany (F.H.P.); Fetal Medicine Unit, St Georges University Hospital Foundation NHS Trust and St Georges University London, UK (B.T., A.K.); and HealthTwiSt GmbH, Berlin, Germany (A.B.)
| | - Baskaran Thilaganathan
- From the Departments of Obstetrics (S.V., L.A.D., W.H.) and Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry (F.H.P.), Charité Universitätsmedizin Berlin, Germany; Labor Berlin - Charité Vivantes GmbH, Germany (F.H.P.); Fetal Medicine Unit, St Georges University Hospital Foundation NHS Trust and St Georges University London, UK (B.T., A.K.); and HealthTwiSt GmbH, Berlin, Germany (A.B.)
| | - Lisa Antonia Dröge
- From the Departments of Obstetrics (S.V., L.A.D., W.H.) and Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry (F.H.P.), Charité Universitätsmedizin Berlin, Germany; Labor Berlin - Charité Vivantes GmbH, Germany (F.H.P.); Fetal Medicine Unit, St Georges University Hospital Foundation NHS Trust and St Georges University London, UK (B.T., A.K.); and HealthTwiSt GmbH, Berlin, Germany (A.B.)
| | - Wolfgang Henrich
- From the Departments of Obstetrics (S.V., L.A.D., W.H.) and Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry (F.H.P.), Charité Universitätsmedizin Berlin, Germany; Labor Berlin - Charité Vivantes GmbH, Germany (F.H.P.); Fetal Medicine Unit, St Georges University Hospital Foundation NHS Trust and St Georges University London, UK (B.T., A.K.); and HealthTwiSt GmbH, Berlin, Germany (A.B.)
| | - Andreas Busjahn
- From the Departments of Obstetrics (S.V., L.A.D., W.H.) and Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry (F.H.P.), Charité Universitätsmedizin Berlin, Germany; Labor Berlin - Charité Vivantes GmbH, Germany (F.H.P.); Fetal Medicine Unit, St Georges University Hospital Foundation NHS Trust and St Georges University London, UK (B.T., A.K.); and HealthTwiSt GmbH, Berlin, Germany (A.B.)
| | - Asma Khalil
- From the Departments of Obstetrics (S.V., L.A.D., W.H.) and Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry (F.H.P.), Charité Universitätsmedizin Berlin, Germany; Labor Berlin - Charité Vivantes GmbH, Germany (F.H.P.); Fetal Medicine Unit, St Georges University Hospital Foundation NHS Trust and St Georges University London, UK (B.T., A.K.); and HealthTwiSt GmbH, Berlin, Germany (A.B.)
| |
Collapse
|
981
|
DLX3 interacts with GCM1 and inhibits its transactivation-stimulating activity in a homeodomain-dependent manner in human trophoblast-derived cells. Sci Rep 2017; 7:2009. [PMID: 28515447 PMCID: PMC5435702 DOI: 10.1038/s41598-017-02120-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 04/06/2017] [Indexed: 11/25/2022] Open
Abstract
The placental transcription factors Distal-less 3 (DLX3) and Glial cell missing-1 (GCM1) have been shown to coordinate the specific regulation of PGF in human trophoblast cell lines. While both factors independently have a positive effect on PGF gene expression, when combined, DLX3 acts as an antagonist to GCM. Despite this understanding, potential mechanisms accounting for this regulatory interaction remain unexplored. We identify physical and functional interactions between specific domains of DLX3 and GCM1 in human trophoblast-derived cells by performing immunoprecipitation and mammalian one hybrid assays. Studies revealed that DLX3 binding reduced the transcriptional activity of GCM1, providing a mechanistic explanation of their functional antagonism in regulating PGF promoter activity. The DLX3 homeodomain (HD) was essential for DLX3-GCM1 interaction, and that the HD together with the DLX3 amino- or carboxyl-terminal domains was required for maximal inhibition of GCM1. Interestingly, a naturally occurring DLX3 mutant that disrupts the carboxyl-terminal domain leading to tricho-dento-osseous syndrome in humans displayed activities indistinguishable from wild type DLX3 in this system. Collectively, our studies demonstrate that DLX3 physically interacts with GCM1 and inhibits its transactivation activity, suggesting that DLX3 and GCM1 may form a complex to functionally regulate placental cell function through modulation of target gene expression.
Collapse
|
982
|
Abstract
OPINION STATEMENT Cardiovascular disease (CVD) is the leading cause of pregnancy-associated mortality, with an increasingly complex pregnant population. While our understanding of CVD in pregnancy continues to evolve, there remains a need to develop widely accessible tools to follow pregnant women both with and without preexisting disease with respect to cardiovascular risk, particularly for those presenting with symptoms suggestive of cardiovascular pathology. Thus, research is emerging with respect to the potential role of novel and established cardiac biomarkers in diagnosing and following CVD in pregnancy. Here, we review the normal hemodynamics of pregnancy and the behavior of various biomarkers in both normal and complicated pregnancies.
Collapse
Affiliation(s)
- Emily S Lau
- Division of Cardiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA
| | - Amy Sarma
- Division of Cardiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA.
| |
Collapse
|
983
|
Abstract
OBJECTIVE To evaluate the risk of preeclampsia in pregnant women with human immunodeficiency virus (HIV). METHODS This is a 26-year population-based retrospective cohort study. Human immunodeficiency virus-infected pregnant women were compared with a HIV-negative comparison group. The primary outcome was the incidence of preeclampsia. We planned subgroup analysis according to antiretroviral therapy. RESULTS A total of 84,725 women were included in the analysis, of whom 453 were HIV-infected and 84,272 HIV-negative. Of the 453 HIV-infected women, 301 (66.4%) received highly active antiretroviral therapy (HAART group) during pregnancy, whereas 152 (33.6%) did not. After adjusting for confounders, we found that HIV-infected women had a significantly higher risk of preeclampsia (10.2% compared with 4.1%; adjusted odds ratio [OR] 2.68, 95% confidence interval [CI] 1.96-3.64), preeclampsia with severe features (4.0% compared with 2.0%; adjusted OR 2.03, 95% CI 1.26-3.28), early-onset (3.5% compared with 1.4%; adjusted OR 2.50, 95% CI 1.51-4.15) and late-onset preeclampsia (6.6% compared with 2.6%; adjusted OR 2.64, 95% CI 1.82-3.85), and preterm birth at less than 37 weeks of gestation (11.0% compared with 4.7%; adjusted OR 2.50, 95% CI 1.86-3.37) compared with the comparison group. Human immunodeficiency virus-infected women who received HAART had a significantly higher risk of preeclampsia compared with women without HIV (13.0% compared with 4.1%; adjusted OR 3.52, 95% CI 2.51-4.94) and compared with the non-HAART group (13.0% compared with 4.6%; adjusted OR 3.08, 95% CI 1.34-5.07). The non-HAART group had a similar risk compared with women without HIV (4.6% compared with 4.1%; adjusted OR 1.14, 95% CI 0.53-2.44). CONCLUSION Human immunodeficiency virus-infected women had an increased risk of preeclampsia. Some of this risk seems to be linked to HAART.
Collapse
|
984
|
Montagnana M, Danese E, Lippi G, Fava C. Blood laboratory testing for early prediction of preeclampsia: chasing the finish line or at the starting blocks? Ann Med 2017; 49:240-253. [PMID: 27791388 DOI: 10.1080/07853890.2016.1255350] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Preeclampsia (PE) affects 2-8% of pregnancies worldwide, thus representing an important cause of maternal and neonatal morbidity, up to death. Many studies have been designed to identify putative biomarkers for accurate and timely diagnosing PE, but only some of them were focused on specific and sensitive biomarkers for early prediction of this life-threatening condition. In particular, some prospective studies aimed to investigate the predictive role of circulating biomarkers before 20 weeks of gestation in the general pregnant population yielded conflicting results. This article is hence centered on results obtained in studies investigating the predictive performances of angiogenic, anti-angiogenic, inflammatory, endocrine, and epigenetic biomarkers. The available evidence suggests that angiogenic and anti-angiogenic molecules, in particular the sFlt1:PlGF ratio, may be considered the biomarkers with the best diagnostic performance in the second trimester. However, doubts remain about their use in clinical settings before the 20th gestational week. Even lower evidence is available for other biomarkers, due to the fact that some positive results have not been confirmed in ensuing investigations, whereas unresolved analytical issues still contribute to make their clinical reliability rather questionable. Differential expression of microRNAs seems also a promising evidence for early prediction of PE, but additional research and well-designed prospective studies are needed to identify and validate routine predictive tests. KEY MESSAGES Preeclampsia affects 2-8% of pregnant women worldwide, thus remaining one of the leading causes of maternal and neonatal morbidity and mortality. Several studies have investigated the predictive role of circulating biomarkers before 20th week of gestation with conflicting results. Additional research and well-designed prospective studies are needed to identify and validate predictive tests in clinical practice.
Collapse
Affiliation(s)
- Martina Montagnana
- a Sezione di Biochimica Clinica, Dipartimento di Neuroscienze , Biomedicina e Movimento Università di Verona , Italy
| | - Elisa Danese
- a Sezione di Biochimica Clinica, Dipartimento di Neuroscienze , Biomedicina e Movimento Università di Verona , Italy
| | - Giuseppe Lippi
- a Sezione di Biochimica Clinica, Dipartimento di Neuroscienze , Biomedicina e Movimento Università di Verona , Italy
| | - Cristiano Fava
- b Sezione di Medicina Interna C, Dipartimento di Medicina , Università di Verona , Italy
| |
Collapse
|
985
|
Akolekar R. Reply. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2017; 49:666-667. [PMID: 28471028 DOI: 10.1002/uog.17453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Affiliation(s)
- R Akolekar
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, UK
- Department of Fetal Medicine, Medway Maritime Hospital, Gillingham, UK
| |
Collapse
|
986
|
Vatish M, Stepan H, Hund M, Verlohren S. Re: Screening for pre-eclampsia using sFlt-1/PlGF ratio cut-off of 38 at 30-37 weeks' gestation. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2017; 49:665-666. [PMID: 28471023 DOI: 10.1002/uog.17451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 11/14/2016] [Indexed: 06/07/2023]
Affiliation(s)
- M Vatish
- University of Oxford, Oxford, UK
| | - H Stepan
- University of Leipzig, Leipzig, Germany
| | - M Hund
- Roche Diagnostics International, Rotkreuz, Switzerland
| | - S Verlohren
- Charité University Medicine, Berlin, Germany
| |
Collapse
|
987
|
Piccoli GB, Cabiddu G, Castellino S, Gernone G, Santoro D, Moroni G, Spotti D, Giacchino F, Attini R, Limardo M, Maxia S, Fois A, Gammaro L, Todros T. A best practice position statement on the role of the nephrologist in the prevention and follow-up of preeclampsia: the Italian study group on kidney and pregnancy. J Nephrol 2017; 30:307-317. [PMID: 28434090 DOI: 10.1007/s40620-017-0390-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/09/2017] [Indexed: 02/07/2023]
Abstract
Preeclampsia (PE) is a protean syndrome causing a transitory kidney disease, characterised by hypertension and proteinuria, ultimately reversible after delivery. Its prevalence is variously estimated, from 3 to 5% to 10% if all the related disorders, including also pregnancy-induced hypertension (PIH) and HELLP syndrome (haemolysis, increase in liver enzyme, low platelets) are included. Both nephrologists and obstetricians are involved in the management of the disease, according to different protocols, and the clinical management, as well as the role for each specialty, differs worldwide. The increased awareness of the role of chronic kidney disease in pregnancy, complicating up to 3% of pregnancies, and the knowledge that PE is associated with an increased risk for development of CKD later in life have recently increased the interest and redesigned the role of the nephrologists in this context. However, while the heterogeneous definitions of PE, its recent reclassification, an emerging role for biochemical biomarkers, the growing body of epidemiological data and the new potential therapeutic interventions lead to counsel long-term follow-up, the lack of resources for chronic patients and the increasing costs of care limit the potential for preventive actions, and suggest tailoring specific interventional strategies. The aim of the present position statement of the Kidney and Pregnancy Study Group of the Italian Society of Nephrology is to review the literature and to try to identify theoretical and pragmatic bases for an agreed management of PE in the nephrological setting, with particular attention to the prevention of the syndrome (recurrent PE, presence of baseline CKD) and to the organization of the postpartum follow-up.
Collapse
Affiliation(s)
- Giorgina Barbara Piccoli
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy. .,Nephrologie, Centre Hospitalier Le Mans, Avenue Roubillard, 72000, Le Mans, France.
| | | | | | | | | | - Gabriella Moroni
- Nephrology, Fondazione Ca' Granda Ospedale Maggiore, Milan, Italy
| | - Donatella Spotti
- Nephrology and Dialysis, IRCCS Ospedale San Raffaele, Milano, Italy
| | | | - Rossella Attini
- Obstetrics, Department of Surgery, University of Torino, Turin, Italy
| | - Monica Limardo
- Nephrology, Azienda Ospedaliera della Provincia di Lecco, Lecco, Italy
| | | | - Antioco Fois
- Nephrology, Azienda Ospedaliera Brotzu, Cagliari, Italy
| | - Linda Gammaro
- Nephrology Ospedale Fracastoro San Bonifacio, San Bonifacio, Italy
| | - Tullia Todros
- Obstetrics, Department of Surgery, University of Torino, Turin, Italy
| | | |
Collapse
|
988
|
Boyd HA, Basit S, Behrens I, Leirgul E, Bundgaard H, Wohlfahrt J, Melbye M, Øyen N. Association Between Fetal Congenital Heart Defects and Maternal Risk of Hypertensive Disorders of Pregnancy in the Same Pregnancy and Across Pregnancies. Circulation 2017; 136:39-48. [PMID: 28424221 DOI: 10.1161/circulationaha.116.024600] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 04/12/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Both pregnant women carrying fetuses with heart defects and women with hypertensive disorders of pregnancy often exhibit angiogenic imbalances, suggesting that the same mechanisms are involved in the pathogenesis of the former and the pathophysiology of the latter. We conducted a register-based cohort study to determine whether offspring congenital heart defects are associated with an increased risk of hypertensive disorders of pregnancy and whether the mechanisms driving any association are primarily maternal or fetal. METHODS Among singleton pregnancies without chromosomal abnormalities lasting ≥20 weeks in Denmark from 1978 to 2011 (n= 1 972 857), we identified pregnancies complicated by offspring congenital heart defects or early preterm preeclampsia, late preterm preeclampsia, term preeclampsia, and gestational hypertension. We used polytomous logistic regression to estimate odds ratios (ORs) for associations between offspring congenital heart defects and maternal hypertensive disorders of pregnancy overall and for specific heart defects. RESULTS Offspring congenital heart defects were strongly associated with early preterm preeclampsia (OR, 7.00; 95% confidence interval [CI], 6.11-8.03) and late preterm preeclampsia (OR, 2.82; 95% CI, 2.38-3.34) in the same pregnancy and weakly associated with term preeclampsia (OR, 1.16; 95% CI, 1.06-1.27), but they were not associated with gestational hypertension (OR, 1.07; 95% CI, 0.92-1.25). Association strengths were consistent across heart defect types. Offspring congenital heart defects in a previous pregnancy were also strongly associated with preterm preeclampsia in subsequent pregnancies (early preterm preeclampsia: OR, 2.37; 95% CI, 1.68-3.34; late preterm preeclampsia: OR, 2.04; 95% CI, 1.52-2.75) but were only modestly associated with term preeclampsia and not associated with gestational hypertension. Similarly, preterm preeclampsia in a previous pregnancy, but not term preeclampsia or gestational hypertension, was associated with offspring congenital heart defects in later pregnancies (early preterm preeclampsia: OR, 7.91; 95% CI, 6.06-10.3; late preterm preeclampsia: OR, 2.83; 95% CI, 2.11-3.79; term preeclampsia: OR, 0.98; 95% CI, 0.88-1.10; gestational hypertension: OR, 1.13; 95% CI, 0.92-1.38). CONCLUSIONS Linked pathophysiological mechanisms may be involved in some congenital heart defects and preterm preeclampsia. The strong associations across pregnancies support a predominantly maternal origin of effect.
Collapse
Affiliation(s)
- Heather Allison Boyd
- From Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark (H.A.B., S.B., I.B., J.W., M.M.); Department of Global Public Health and Primary Care, University of Bergen, Norway (E.L., N.Ø.); Department of Cardiology (E.L.) and Center for Medical Genetics and Molecular Medicine (N.Ø.), Haukeland University Hospital, Bergen, Norway; Unit for Inherited Cardiac Diseases, Heart Centre, Copenhagen University Hospital (Rigshospitalet), Denmark (H.B.); Department of Clinical Medicine, Copenhagen University, Denmark (M.M.); and Department of Medicine, Stanford University School of Medicine, CA (M.M.).
| | - Saima Basit
- From Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark (H.A.B., S.B., I.B., J.W., M.M.); Department of Global Public Health and Primary Care, University of Bergen, Norway (E.L., N.Ø.); Department of Cardiology (E.L.) and Center for Medical Genetics and Molecular Medicine (N.Ø.), Haukeland University Hospital, Bergen, Norway; Unit for Inherited Cardiac Diseases, Heart Centre, Copenhagen University Hospital (Rigshospitalet), Denmark (H.B.); Department of Clinical Medicine, Copenhagen University, Denmark (M.M.); and Department of Medicine, Stanford University School of Medicine, CA (M.M.)
| | - Ida Behrens
- From Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark (H.A.B., S.B., I.B., J.W., M.M.); Department of Global Public Health and Primary Care, University of Bergen, Norway (E.L., N.Ø.); Department of Cardiology (E.L.) and Center for Medical Genetics and Molecular Medicine (N.Ø.), Haukeland University Hospital, Bergen, Norway; Unit for Inherited Cardiac Diseases, Heart Centre, Copenhagen University Hospital (Rigshospitalet), Denmark (H.B.); Department of Clinical Medicine, Copenhagen University, Denmark (M.M.); and Department of Medicine, Stanford University School of Medicine, CA (M.M.)
| | - Elisabeth Leirgul
- From Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark (H.A.B., S.B., I.B., J.W., M.M.); Department of Global Public Health and Primary Care, University of Bergen, Norway (E.L., N.Ø.); Department of Cardiology (E.L.) and Center for Medical Genetics and Molecular Medicine (N.Ø.), Haukeland University Hospital, Bergen, Norway; Unit for Inherited Cardiac Diseases, Heart Centre, Copenhagen University Hospital (Rigshospitalet), Denmark (H.B.); Department of Clinical Medicine, Copenhagen University, Denmark (M.M.); and Department of Medicine, Stanford University School of Medicine, CA (M.M.)
| | - Henning Bundgaard
- From Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark (H.A.B., S.B., I.B., J.W., M.M.); Department of Global Public Health and Primary Care, University of Bergen, Norway (E.L., N.Ø.); Department of Cardiology (E.L.) and Center for Medical Genetics and Molecular Medicine (N.Ø.), Haukeland University Hospital, Bergen, Norway; Unit for Inherited Cardiac Diseases, Heart Centre, Copenhagen University Hospital (Rigshospitalet), Denmark (H.B.); Department of Clinical Medicine, Copenhagen University, Denmark (M.M.); and Department of Medicine, Stanford University School of Medicine, CA (M.M.)
| | - Jan Wohlfahrt
- From Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark (H.A.B., S.B., I.B., J.W., M.M.); Department of Global Public Health and Primary Care, University of Bergen, Norway (E.L., N.Ø.); Department of Cardiology (E.L.) and Center for Medical Genetics and Molecular Medicine (N.Ø.), Haukeland University Hospital, Bergen, Norway; Unit for Inherited Cardiac Diseases, Heart Centre, Copenhagen University Hospital (Rigshospitalet), Denmark (H.B.); Department of Clinical Medicine, Copenhagen University, Denmark (M.M.); and Department of Medicine, Stanford University School of Medicine, CA (M.M.)
| | - Mads Melbye
- From Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark (H.A.B., S.B., I.B., J.W., M.M.); Department of Global Public Health and Primary Care, University of Bergen, Norway (E.L., N.Ø.); Department of Cardiology (E.L.) and Center for Medical Genetics and Molecular Medicine (N.Ø.), Haukeland University Hospital, Bergen, Norway; Unit for Inherited Cardiac Diseases, Heart Centre, Copenhagen University Hospital (Rigshospitalet), Denmark (H.B.); Department of Clinical Medicine, Copenhagen University, Denmark (M.M.); and Department of Medicine, Stanford University School of Medicine, CA (M.M.)
| | - Nina Øyen
- From Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark (H.A.B., S.B., I.B., J.W., M.M.); Department of Global Public Health and Primary Care, University of Bergen, Norway (E.L., N.Ø.); Department of Cardiology (E.L.) and Center for Medical Genetics and Molecular Medicine (N.Ø.), Haukeland University Hospital, Bergen, Norway; Unit for Inherited Cardiac Diseases, Heart Centre, Copenhagen University Hospital (Rigshospitalet), Denmark (H.B.); Department of Clinical Medicine, Copenhagen University, Denmark (M.M.); and Department of Medicine, Stanford University School of Medicine, CA (M.M.)
| |
Collapse
|
989
|
Porreco RP, Heyborne KD. Immunogenesis of preeclampsia: lessons from donor gametes. J Matern Fetal Neonatal Med 2017; 31:1220-1226. [DOI: 10.1080/14767058.2017.1309385] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Richard P. Porreco
- Maternal Fetal Medicine, Presbyterian/St.Luke’s Medical Center, Obstetrix Medical Group of Colorado, Denver, CO, USA
| | - Kent D. Heyborne
- Obstetrics and Gynecology, Denver Health and Hospital Authority, Denver, CO, USA
| |
Collapse
|
990
|
Surányi A, Altorjay Á, Kaiser L, Nyári T, Németh G. Evaluation of placental vascularization by three-dimensional ultrasound examination in second and third trimester of pregnancies complicated by chronic hypertension, gestational hypertension or pre-eclampsia. Pregnancy Hypertens 2017; 8:51-59. [DOI: 10.1016/j.preghy.2017.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 01/27/2017] [Accepted: 03/17/2017] [Indexed: 10/19/2022]
|
991
|
Dröge LA, Höller A, Ehrlich L, Verlohren S, Henrich W, Perschel FH. Diagnosis of preeclampsia and fetal growth restriction with the sFlt-1/PlGF ratio: Diagnostic accuracy of the automated immunoassay Kryptor®. Pregnancy Hypertens 2017; 8:31-36. [DOI: 10.1016/j.preghy.2017.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 02/05/2017] [Accepted: 02/27/2017] [Indexed: 11/25/2022]
|
992
|
Stallone G, Matteo M, Netti GS, Infante B, Di Lorenzo A, Prattichizzo C, Carlucci S, Trezza F, Gesualdo L, Greco P, Grandaliano G. Semaphorin 3F expression is reduced in pregnancy complicated by preeclampsia. An observational clinical study. PLoS One 2017; 12:e0174400. [PMID: 28350837 PMCID: PMC5370113 DOI: 10.1371/journal.pone.0174400] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 03/08/2017] [Indexed: 12/22/2022] Open
Abstract
Background and objective Preeclampsia is a systemic disorder, affecting 2–10% of pregnancies, characterized by a deregulated pro- and anti-angiogenic balance. Semaphorin 3F is an angiogenesis inhibitor. We aimed to investigate whether semaphorin 3F expression is modulated in preeclampsia. Design, setting, participants, and measurements We performed two observational single center cohort studies between March 2013 and August 2014. In the first we enrolled 110 consecutive women, undergoing an elective cesarean section; in the second we included 150 consecutive women undergoing amniocentesis for routine clinical indications at 16–18 week of gestation. Semaphorin 3F concentration was evaluated in maternal peripheral blood, venous umbilical blood and amniotic fluid, along with its placenta protein expression at the time of delivery in the first study group and in the amniotic fluid at 16–18 weeks of gestation in the second study group. Results In the first study 19 patients presented at delivery with preeclampsia. Semaphorin 3F placenta tissue expression was significantly reduced in preeclampsia. In addition, semaphorin 3F level at delivery was significantly lower in serum, amniotic fluid and venous umbilical blood of preeclamptic patients compared with normal pregnant women. In the prospective cohort study 14 women developed preeclampsia. In this setting, semaphorin 3F amniotic level at 16–18 weeks of gestation was reduced in women who subsequently developed preeclampsia compared to women with a normal pregnancy. ROC curve analysis showed that semaphorin 3F amniotic levels could identify women at higher risk of preeclampsia. Conclusions Semaphorin 3F might represent a predictive biomarker of preeclampsia.
Collapse
Affiliation(s)
- Giovanni Stallone
- Nephrology Dialysis and Transplantation Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Maria Matteo
- Gynaecologic and Obstetric Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Giuseppe Stefano Netti
- Clinical Pathology Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Barbara Infante
- Nephrology Dialysis and Transplantation Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Adelaide Di Lorenzo
- Nephrology Dialysis and Transplantation Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Clelia Prattichizzo
- Nephrology Dialysis and Transplantation Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Stefania Carlucci
- Gynaecologic and Obstetric Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Federica Trezza
- Gynaecologic and Obstetric Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Loreto Gesualdo
- Nephrology Dialysis and Transplantation Unit, Dept. of Emergency and Organ Transplantation, University of Bari “A. Moro”, Piazza G. Cesare 11, Bari, Italy
| | - Pantaleo Greco
- Gynaecologic and Obstetric Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Giuseppe Grandaliano
- Nephrology Dialysis and Transplantation Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
- * E-mail:
| |
Collapse
|
993
|
Abstract
Hypertension and chronic kidney disease (CKD) have a significant impact on global morbidity and mortality. The Low Birth Weight and Nephron Number Working Group has prepared a consensus document aimed to address the relatively neglected issue for the developmental programming of hypertension and CKD. It emerged from a workshop held on April 2, 2016, including eminent internationally recognized experts in the field of obstetrics, neonatology, and nephrology. Through multidisciplinary engagement, the goal of the workshop was to highlight the association between fetal and childhood development and an increased risk of adult diseases, focusing on hypertension and CKD, and to suggest possible practical solutions for the future. The recommendations for action of the consensus workshop are the results of combined clinical experience, shared research expertise, and a review of the literature. They highlight the need to act early to prevent CKD and other related noncommunicable diseases later in life by reducing low birth weight, small for gestational age, prematurity, and low nephron numbers at birth through coordinated interventions. Meeting the current unmet needs would help to define the most cost-effective strategies and to optimize interventions to limit or interrupt the developmental programming cycle of CKD later in life, especially in the poorest part of the world.
Collapse
|
994
|
Zhao M, Zhu Z, Liu C, Zhang Z. Dual-cutoff of sFlt-1/PlGF ratio in the stratification of preeclampsia: a systematic review and meta-analysis. Arch Gynecol Obstet 2017; 295:1079-1087. [PMID: 28314983 DOI: 10.1007/s00404-017-4302-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/13/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE To systematically review the approach of using two independent sFlt-1/PlGF cutoffs that has better sensitivity (cutoff-sen) and specificity (cutoff-spe) separately for risk stratification in the detection of preeclampsia. METHODS PubMed and Embase databases and reference lists were searched up to June 2016. Inclusion criteria were blood samples for sFlt-1/PlGF with separate cutoffs (cutoff-sen and cutoff-spe) provided. Six relevant studies were identified. Pooling of results was done based on three studies and a systematic review was performed based on all six. RESULTS The strategy of using a cutoff of ≤33 and ≥85 for early onset preeclampsia, and ≤33 and ≥110 for the late onset preeclampsia was proposed and examined. The pooled sensitivity for cutoff-sen was: 95.3% (90.6-98.1%) and 88.6% (82.9-92.9%) for early and late onset preeclampsia, respectively. The pooled specificity for cutoff-spe was: 97.6% (95.2-98.9%) and 94.2% (91.4-96.3%) for early and late onset preeclampsia respectively. The pooled estimation of the early onset pre-eclamptic pregnancies and control normal pregnancies classified in the equivocal zone was 4.9% (2.0-8.8%) and 32.4% (25.7-39.5%), respectively, and 26.8% (10.3-47.6%) and 8.7% (3.0-17.6%) for late onset patients. CONCLUSION The new dual-cutoff diagnostic system optimizes the predictive performance of the single cutoff system. Further studies are required to assess the performance of this system and to define the approach and frequency at which subjects in the equivocal zone should be screened.
Collapse
Affiliation(s)
- Mutong Zhao
- Department of Obstetrics and Gynecology, Beijng Chao-yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Zhiqing Zhu
- Department of Obstetrics and Gynecology, Beijng Chao-yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Chongdong Liu
- Department of Obstetrics and Gynecology, Beijng Chao-yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Zhenyu Zhang
- Department of Obstetrics and Gynecology, Beijng Chao-yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China.
| |
Collapse
|
995
|
Ratio of matrix metalloproteinase-2 to -9 is a more accurate predictive biomarker in women with suspected pre-eclampsia. Biosci Rep 2017; 37:BSR20160508. [PMID: 28143958 PMCID: PMC5350602 DOI: 10.1042/bsr20160508] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 01/21/2017] [Accepted: 01/31/2017] [Indexed: 11/29/2022] Open
Abstract
Pre-eclampsia (PE) is a condition unique to pregnancy, and abnormal expression of matrix metalloproteinases (MMPs) has been implicated in its pathogenesis. We aimed to evaluate the reliability of plasma levels of MMP-2, MMP-9 and their relative ratio in predicting PE. A total of 318 women with suspected PE were recruited for the study, who were subsequently either cleared or diagnosed of PE and grouped accordingly. Their baseline characteristics were compared. Blood samples were also collected from all participants, to determine the plasma levels of MMP-2 and MMP-9. The predictive values of levels of MMP-2 and MMP-9, as well as their ratio, were analyzed using the receiver operating characteristic (ROC) curve. Either MMP-2 or MMP-9 alone did not exhibit any obvious differences between normal and PE pregnancies. However the ratio of MMP-2/MMP-9 was significantly higher in PE-affected pregnancy than normal control group. ROC curve analysis also indicated that the MMP-2/MMP-9 ratio provided better compromise between specificity and sensitivity in distinguishing PE from normal pregnancies, than either of the two MMPs alone. MMP-2/MMP-9 ratio is a more accurate biomarker to predict PE than either MMP-2 or MMP-9 alone.
Collapse
|
996
|
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
|
997
|
Ruiz A, Cruz-Lemini M, Masoller N, Sanz-Cortés M, Ferrer Q, Ribera I, Martínez JM, Crispi F, Arévalo S, Gómez O, Pérez-Hoyos S, Carreras E, Gratacós E, Llurba E. Longitudinal changes in fetal biometry and cerebroplacental hemodynamics in fetuses with congenital heart disease. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2017; 49:379-386. [PMID: 27214694 DOI: 10.1002/uog.15970] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 04/20/2016] [Accepted: 04/29/2016] [Indexed: 06/05/2023]
Abstract
OBJECTIVES To determine the longitudinal behavior of fetal biometric measures and cerebroplacental hemodynamics throughout gestation in fetuses with congenital heart disease (CHD). METHODS Fetal biometry and Doppler hemodynamics (uterine artery (UtA), umbilical artery (UA) and fetal middle cerebral artery (MCA)) were measured serially in a cohort of consecutive fetuses diagnosed with CHD. Evaluations were made at various time points, from diagnosis (20-25 weeks) to delivery, with at least two measurements per fetus that were at least 2 weeks apart. Fetuses were classified into three groups according to the pattern of blood supply to the brain (placental vs systemic) that would be expected on the basis of the type of CHD. All parameters were transformed into Z-scores. A linear mixed model to analyze repeated measurements was constructed for each parameter to assess its behavior throughout gestation. RESULTS Four hundred and forty-four ultrasound examinations were performed in 119 CHD fetuses, with a median of two measurements per fetus. The fetuses presented a small head at diagnosis (biparietal diameter (BPD) Z-score, -1.32 ± 0.99; head circumference (HC) Z-score, -0.79 ± 1.02), which remained small throughout gestation. UtA and UA pulsatility indices (PI) showed a significant increase towards the end of pregnancy, whereas no significant changes were observed in MCA-PI or cerebroplacental ratio (CPR) with gestational age. Both MCA and CPR presented significant differences in longitudinal behavior between CHD groups, while BPD and HC did not. CONCLUSIONS CHD fetuses have a relatively small head from the second trimester of pregnancy, regardless of the type of CHD anomaly, and increasing resistance in the UtA and UA as pregnancy progresses, suggestive of increasing degree of placental impairment. Our findings indicate the early onset of mechanisms that could lead to poorer neurodevelopment later in life. Copyright © 2016 ISUOG. Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- A Ruiz
- Department of Obstetrics, Maternal-Fetal Medicine Unit, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
- Maternal and Child Health and Development Network II (SAMID II) RD12/0026, Institute of Health Carlos III, Madrid, Spain
| | - M Cruz-Lemini
- Department of Obstetrics, Maternal-Fetal Medicine Unit, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
- Maternal and Child Health and Development Network II (SAMID II) RD12/0026, Institute of Health Carlos III, Madrid, Spain
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
- Fetal Medicine Mexico, Fetal Medicine and Surgery Research Unit, Unidad de Investigación en Neurodesarrollo, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM) Campus Juriquilla, Querétaro, Mexico
| | - N Masoller
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - M Sanz-Cortés
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
- Maternal Fetal Medicine Department, Baylor College of Medicine, Houston, TX, USA
| | - Q Ferrer
- Department of Obstetrics, Maternal-Fetal Medicine Unit, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
- Maternal and Child Health and Development Network II (SAMID II) RD12/0026, Institute of Health Carlos III, Madrid, Spain
| | - I Ribera
- Department of Obstetrics, Maternal-Fetal Medicine Unit, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
- Maternal and Child Health and Development Network II (SAMID II) RD12/0026, Institute of Health Carlos III, Madrid, Spain
| | - J M Martínez
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - F Crispi
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - S Arévalo
- Department of Obstetrics, Maternal-Fetal Medicine Unit, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
- Maternal and Child Health and Development Network II (SAMID II) RD12/0026, Institute of Health Carlos III, Madrid, Spain
| | - O Gómez
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - S Pérez-Hoyos
- Department of Statistics and Preventive Medicine, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - E Carreras
- Department of Obstetrics, Maternal-Fetal Medicine Unit, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
- Maternal and Child Health and Development Network II (SAMID II) RD12/0026, Institute of Health Carlos III, Madrid, Spain
| | - E Gratacós
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - E Llurba
- Department of Obstetrics, Maternal-Fetal Medicine Unit, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
- Maternal and Child Health and Development Network II (SAMID II) RD12/0026, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
998
|
Li S, Roberson MS. Dlx3 and GCM-1 functionally coordinate the regulation of placental growth factor in human trophoblast-derived cells. J Cell Physiol 2017; 232:2900-2914. [PMID: 27996093 DOI: 10.1002/jcp.25752] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/19/2016] [Accepted: 12/19/2016] [Indexed: 11/08/2022]
Abstract
Placental growth factor (PGF) is abundantly expressed by trophoblast cells within human placentae and is important for trophoblast development and placental vascularization. Circulating maternal serum levels of PGF are dynamically upregulated across gestation in normal pregnancies, whereas low circulating levels and placental production of PGF have been implicated in the pathogenesis of preeclampsia and other gestational diseases. However, the underlying molecular mechanism of regulating PGF expression in the human placenta remains poorly understood. In this study, we demonstrated that transcription factors Distal-less 3 (DLX3) and Glial cell missing-1 (GCM1) were both sufficient and required for PGF expression in human trophoblast-derived cells by overexpression and knockdown approaches. Surprisingly, while DLX3 and GCM1 were both positive regulators of PGF, co-overexpression of DLX3 and GCM1 led to an antagonist effect on PGF expression on the endogenous gene and a luciferase reporter. Further, deletion and site-directed mutagenesis studies identified a novel regulatory element on the PGF promoter mediating both DLX3- and GCM1-dependent PGF expression. This regulatory region was also found to be essential for the basal activity of the PGF promoter. Finally, Chromatin-immunoprecipitation (ChIP) assays revealed colocalization of DLX3 and GCM1 at the identified regulatory region on the PGF promoter. Taken together, our studies provide important insights into intrinsic regulation of human placental PGF expression through the functional coordination of DLX3 and GCM1, and are likely to further the understanding of pathogenesis of PGF dysregulation in preeclampsia and other disease conditions.
Collapse
Affiliation(s)
- Sha Li
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - Mark S Roberson
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York
| |
Collapse
|
999
|
Rhee C, Edwards M, Dang C, Harris J, Brown M, Kim J, Tucker HO. ARID3A is required for mammalian placenta development. Dev Biol 2017; 422:83-91. [PMID: 27965054 PMCID: PMC5540318 DOI: 10.1016/j.ydbio.2016.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 11/29/2016] [Accepted: 12/01/2016] [Indexed: 11/17/2022]
Abstract
Previous studies in the mouse indicated that ARID3A plays a critical role in the first cell fate decision required for generation of trophectoderm (TE). Here, we demonstrate that ARID3A is widely expressed during mouse and human placentation and essential for early embryonic viability. ARID3A localizes to trophoblast giant cells and other trophoblast-derived cell subtypes in the junctional and labyrinth zones of the placenta. Conventional Arid3a knockout embryos suffer restricted intrauterine growth with severe defects in placental structural organization. Arid3a null placentas show aberrant expression of subtype-specific markers as well as significant alteration in cytokines, chemokines and inflammatory response-related genes, including previously established markers of human placentation disorders. BMP4-mediated induction of trophoblast stem (TS)-like cells from human induced pluripotent stem cells results in ARID3A up-regulation and cytoplasmic to nuclear translocation. Overexpression of ARID3A in BMP4-mediated TS-like cells up-regulates TE markers, whereas pluripotency markers are down-regulated. Our results reveal an essential, conserved function for ARID3A in mammalian placental development through regulation of both intrinsic and extrinsic developmental programs.
Collapse
Affiliation(s)
- Catherine Rhee
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States
| | - Melissa Edwards
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States; Cell and Molecular Biology, Colorado State University, Fort Collins, CO 80523, United States
| | - Christine Dang
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States
| | - June Harris
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States
| | - Mark Brown
- Cell and Molecular Biology, Colorado State University, Fort Collins, CO 80523, United States
| | - Jonghwan Kim
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States
| | - Haley O Tucker
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States.
| |
Collapse
|
1000
|
Hagmann H, Thadhani RI. Angiogenic Markers in Transition: Thinking Positive. Hypertension 2017; 69:578-579. [PMID: 28167684 DOI: 10.1161/hypertensionaha.116.08739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Henning Hagmann
- From the Department II of Internal Medicine and Nephrology and Center for Molecular Medicine Cologne, University of Cologne, Germany (H.H.); and Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston (R.I.T.)
| | - Ravi I Thadhani
- From the Department II of Internal Medicine and Nephrology and Center for Molecular Medicine Cologne, University of Cologne, Germany (H.H.); and Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston (R.I.T.).
| |
Collapse
|