151
|
Lekva T, Michelsen AE, Roland MCP, Norwitz ER, Estensen ME, Olstad OK, Akkouh IA, Henriksen T, Bollerslev J, Aukrust P, Ueland T. Increased ferroptosis in leukocytes from preeclamptic women involving the long non-coding taurine upregulated gene 1 (TUG1). J Intern Med 2024; 295:181-195. [PMID: 37870937 DOI: 10.1111/joim.13732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
BACKGROUND Ferroptosis plays a key role in placental development and physiology, and abnormal ferroptosis has been implicated in trophoblast injury leading to preeclampsia (PE). We hypothesize that leukocytes isolated from PE exhibit increased ferroptosis and that extracellular vesicles contain long non-coding (lnc) RNA/mRNAs that modulate oxidative stress and iron toxicity in vascular endothelial cells. METHODS We measured the expression of key regulators of ferroptosis in leukocytes and extracellular vesicles as well as circulating biomarkers of iron homeostasis and oxidative stress in plasma from women with/without PE at different timepoints during pregnancy. For markers that were dysregulated, we assessed their temporal correlation with established markers of disease activity and marker of endothelial activation. For markers dysregulated in early pregnancy, we assessed their ability to predict the development of PE. RESULTS We found decreased lncRNA/mRNAs in leukocytes, but not extracellular vesicles, in PE that may modulate oxidative stress and iron toxicity. This decrease in anti-ferroptotic markers does not appear to be related to maternal disease activity or plasma oxidative stress status but rather to attenuated anti-inflammatory expression in these cells. Circulating ferritin was elevated in PE, supporting the hypothesis that PE represents a disbalance in iron homeostasis. Low lncRNA taurine upregulated gene 1 RNA levels in leukocytes at 22-24 weeks were strongly associated with the development of PE. CONCLUSIONS Our findings suggest that maternal leukocytes in PE show decreased anti-ferroptotic activity that correlates with anti-inflammatory expression. Moreover, some of these changes in ferroptotic activity appear to precede the development of PE.
Collapse
Affiliation(s)
- Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Annika Elisabet Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - Errol R Norwitz
- Newton-Wellesley Hospital and Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | - Ole Kristoffer Olstad
- The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Ibrahim A Akkouh
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Tore Henriksen
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Obstetrics, Oslo University Hospital, Oslo, Norway
| | - Jens Bollerslev
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
152
|
Mansukhani T, Wright A, Arechvo A, Laich A, Iglesias M, Charakida M, Nicolaides KH. Ophthalmic artery Doppler at 36 weeks' gestation in prediction of pre-eclampsia: validation and update of previous model. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2024; 63:230-236. [PMID: 37616530 DOI: 10.1002/uog.27464] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023]
Abstract
OBJECTIVE To validate and extend a model incorporating maternal ophthalmic artery Doppler at 35-37 weeks' gestation in the prediction of subsequent development of pre-eclampsia (PE). METHODS This was a prospective validation study of screening for PE (defined according to the 2019 American College of Obstetricians and Gynecologists criteria) by maternal ophthalmic artery peak systolic velocity (PSV) ratio in 6746 singleton pregnancies undergoing routine care at 35 + 0 to 36 + 6 weeks' gestation (validation dataset). Additionally, the data from the validation dataset were combined with those of 2287 pregnancies that were previously used for development of the model (training dataset), and the combined data were used to update the original model parameters. The competing-risks model was used to estimate the individual patient-specific risk of delivery with PE at any time and within 3 weeks from assessment by a combination of maternal demographic characteristics and medical history with PSV ratio alone and in combination with the established PE biomarkers of mean arterial pressure (MAP), uterine artery pulsatility index (UtA-PI), serum placental growth factor (PlGF) and serum soluble fms-like tyrosine kinase-1 (sFlt-1). We evaluated the predictive performance of the model by examining, first, the ability to discriminate between the PE and non-PE groups using the area under the receiver-operating-characteristics curve and the detection rate (DR) at fixed screen-positive (SPR) and false-positive rates of 10% and, second, calibration by measuring the calibration slope and calibration-in-the-large. McNemar's test was used to compare the performance of screening by a biophysical test (maternal factors, MAP, UtA-PI and PSV ratio) vs a biochemical test (maternal factors, PlGF and sFlt-1), low PlGF concentration (< 10th percentile) or high sFlt-1/PlGF concentration ratio (> 90th percentile). RESULTS In the validation dataset, the performance of screening by maternal factors and PSV ratio for delivery with PE within 3 weeks and at any time after assessment was consistent with that in the training dataset, and there was good agreement between the predicted and observed incidence of PE. In the combined data from the training and validation datasets, good prediction for PE was achieved in screening by a combination of maternal factors, MAP, UtA-PI, PlGF, sFlt-1 and PSV ratio, with a DR, at a 10% SPR, of 85.0% (95% CI, 76.5-91.4%) for delivery with PE within 3 weeks and 65.7% (95% CI, 59.2-71.7%) for delivery with PE at any time after assessment. The performance of a biophysical test was superior to that of screening by low PlGF concentration or high sFlt-1/PlGF concentration ratio but not significantly different from the performance of a biochemical test combining maternal factors with PlGF and sFlt-1 for both PE within 3 weeks and PE at any time after assessment. CONCLUSION Maternal ophthalmic artery PSV ratio at 35-37 weeks' gestation in combination with other biomarkers provides effective prediction of subsequent development of PE. © 2023 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- T Mansukhani
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, UK
| | - A Wright
- Institute of Health Research, University of Exeter, Exeter, UK
| | - A Arechvo
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, UK
| | - A Laich
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, UK
| | - M Iglesias
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, UK
| | - M Charakida
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, UK
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - K H Nicolaides
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, UK
| |
Collapse
|
153
|
Neave L, Thomas M, de Groot R, Doyle AJ, Singh D, Adams G, David AL, Maksym K, Scully M. Alterations in the von Willebrand factor/ADAMTS-13 axis in preeclampsia. J Thromb Haemost 2024; 22:455-465. [PMID: 37926193 DOI: 10.1016/j.jtha.2023.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/07/2023] [Accepted: 10/25/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Preeclampsia is a gestational hypertensive disorder characterized by maternal endothelial activation and increased ratio of soluble fms-like tyrosine kinase-1 (sFlt-1) inhibitor to placental growth factor (PlGF). The von Willebrand factor (VWF)/ADAMTS-13 axis is of interest because of the underlying endothelial activation and clinical overlap with pregnancy-associated thrombotic thrombocytopenic purpura. OBJECTIVES To assess VWF, ADAMTS-13, and VWF/ADAMTS-13 ratio in preeclampsia and look for associations with sFlt-1/PlGF ratio and clinical features. METHODS Thirty-four preeclampsia cases and 48 normal pregnancies were assessed in a case-control study. Twelve normal pregnancies in women with a history of preeclampsia formed an additional comparator group. VWF antigen (VWF:Ag) and VWF activity (VWF:Ac [VWF:glycoprotein IbM]) were measured via automated immunoturbidimetric assay, ADAMTS-13 activity was measured via fluorescence resonance energy transfer-VWF73 assay, and sFlt-1 and PlGF were measured via enzyme-linked immunosorbent assay. RESULTS VWF:Ag was higher in preeclampsia than in normal pregnancy (median, 3.07 vs 1.87 IU/mL; P < .0001). ADAMTS-13 activity was slightly lower (median, 89.6 vs 94.4 IU/dL; P = .02), with no severe deficiencies. Significant elevations in VWF:Ac were not observed in preeclampsia, resulting in reduced VWF:Ac/VWF:Ag ratios (median, 0.77 vs 0.97; P < .0001). VWF:Ag/ADAMTS-13 ratios were significantly higher in preeclampsia (median, 3.42 vs 2.06; P < .0001), with an adjusted odds ratio of 19.2 for a ratio of >2.7 (>75th centile of normal pregnancy). Those with a history of preeclampsia had similar ratios to normal pregnant controls. VWF:Ag/ADAMTS-13 and sFlt-1/PlGF were not correlated. However, percentage reduction in platelets correlated positively with VWF:Ac (P = .01), VWF:Ac/VWF:Ag ratio (P = .004), and sFlt-1/PlGF ratio (P = .01). CONCLUSION The VWF/ADAMTS-13 axis is significantly altered in preeclampsia. Further investigation of potential clinical utility is warranted.
Collapse
Affiliation(s)
- Lucy Neave
- Department of Clinical Haematology, University College London Hospitals NHS Foundation Trust, London, United Kingdom; Haemostasis Research Unit, University College London, London, United Kingdom.
| | - Mari Thomas
- Department of Clinical Haematology, University College London Hospitals NHS Foundation Trust, London, United Kingdom; National Institute for Health and Care Research University College London Hospital/University College London Biomedical Research Centre, London, United Kingdom
| | - Rens de Groot
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Andrew J Doyle
- Department of Clinical Haematology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Deepak Singh
- Special Coagulation, Health Services Laboratories, London, United Kingdom
| | - George Adams
- Department of Haematology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Anna L David
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, United Kingdom
| | - Katarzyna Maksym
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, United Kingdom
| | - Marie Scully
- Department of Clinical Haematology, University College London Hospitals NHS Foundation Trust, London, United Kingdom; National Institute for Health and Care Research University College London Hospital/University College London Biomedical Research Centre, London, United Kingdom
| |
Collapse
|
154
|
Rajiv P, Cade T, Dean J, Jones GD, Brennecke SP. Maternal serum soluble fms-like tyrosine kinase-1-to-placental growth factor ratio distinguishes growth-restricted from non-growth-restricted small-for-gestational-age fetuses. AJOG GLOBAL REPORTS 2024; 4:100302. [PMID: 38318268 PMCID: PMC10839529 DOI: 10.1016/j.xagr.2023.100302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Fetal growth restriction secondary to chronic placental insufficiency is a major cause of perinatal morbidity and mortality. A significant proportion of fetuses with fetal growth restriction are small for gestational age, defined as a birthweight of ≤10th percentile. However, not all small-for-gestational-age fetuses are growth restricted. Some are constitutionally small and otherwise healthy. It is important to distinguish between small-for-gestational-age fetuses with and without fetal growth restriction to ensure appropriate interventions in small-for-gestational-age fetuses with fetal growth restriction and to minimize unnecessary interventions in healthy small-for-gestational-age fetuses. The maternal serum ratio of soluble fms-like tyrosine kinase-1 and placental growth factor is an indicator of placental insufficiency in the latter half of pregnancy. As such, the soluble fms-like tyrosine kinase-1-to-placental growth factor ratio may be a clinically useful tool to distinguish between small-for-gestational-age fetuses with and without fetal growth restriction. OBJECTIVE This study aimed to determine whether the soluble fms-like tyrosine kinase-1-to-placental growth factor ratio can distinguish between small-for-gestational-age fetuses with and without fetal growth restriction with a birthweight of ≤10th percentile. STUDY DESIGN A retrospective audit of 233 singleton pregnancies delivering an infant with a birthweight of ≤10th percentile corrected for gestational age with an antenatal maternal serum soluble fms-like tyrosine kinase-1-to-placental growth factor result was performed. Fetal growth restriction was defined as a birthweight of ≤10th percentile with an umbilical artery pulsatility index of >95th percentile, fetal middle cerebral artery pulsatility index of <5th percentile, amniotic fluid index of <6 cm, and/or cerebroplacental ratio of <1st percentile. The soluble fms-like tyrosine kinase-1-to-placental growth factor ratios before delivery between fetuses with and without fetal growth restriction (121 [fetal growth restriction] vs 112 [no fetal growth restriction]) were compared. The Student t test and Fisher exact test were used to compare cases and controls. The Mann-Whitney U test, linear regression analysis, and Spearman correlation coefficient (Rho) were used to examine associations between the soluble fms-like tyrosine kinase-1-to-placental growth factor ratio and fetal outcomes to determine whether the soluble fms-like tyrosine kinase-1-to-placental growth factor ratio served as a prognostic marker of fetal growth restriction severity. RESULTS The mean soluble fms-like tyrosine kinase-1-to-placental growth factor ratio was increased in fetal growth restriction cases compared with non-fetal growth restriction controls (234.3±25.0 vs 67.4±7.7, respectively; P<.0001). When controlling for preeclampsia, which is associated with placental insufficiency, fetal growth restriction cases still demonstrated an independent increase in the soluble fms-like tyrosine kinase-1-to-placental growth factor ratio (effect size, 0.865; 95% confidence interval, 0.509-1.220; P<.001). The soluble fms-like tyrosine kinase-1-to-placental growth factor ratio was negatively correlated with birthweight percentiles in pregnancies delivering an infant with a birthweight of ≤10th percentile (r=-0.3565; P<.0001). This association was maintained for fetuses with fetal growth restriction (r=-0.2309; P<.05), whereas fetuses without fetal growth restriction had no significant correlation between the soluble fms-like tyrosine kinase-1-to-placental growth factor ratio and neonatal birthweight percentiles. CONCLUSION The soluble fms-like tyrosine kinase-1-to-placental growth factor ratio was significantly higher in small-for-gestational-age fetuses with fetal growth restriction than small-for-gestational-age fetuses without fetal growth restriction, independent of preeclampsia. Furthermore, the soluble fms-like tyrosine kinase-1-to-placental growth factor ratio was negatively correlated with fetal growth restriction birthweight percentiles, suggesting that it may be a clinical measure of fetal growth restriction severity. Therefore, the ratio may usefully delineate fetal growth restriction from constitutionally small but otherwise healthy fetuses antenatally, allowing for timely interventions in small-for-gestational-age cases with fetal growth restriction and unnecessary interventions to be minimized in small-for-gestational-age cases without fetal growth restriction.
Collapse
Affiliation(s)
- Prithi Rajiv
- Department of Maternal-Fetal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia (Drs Rajiv, Cade, Dean, Davis Jones, and Brennecke)
| | - Thomas Cade
- Department of Maternal-Fetal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia (Drs Rajiv, Cade, Dean, Davis Jones, and Brennecke)
| | - Jennifer Dean
- Department of Maternal-Fetal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia (Drs Rajiv, Cade, Dean, Davis Jones, and Brennecke)
| | - Gabriel Davis Jones
- Department of Maternal-Fetal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia (Drs Rajiv, Cade, Dean, Davis Jones, and Brennecke)
| | - Shaun P. Brennecke
- Department of Maternal-Fetal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia (Drs Rajiv, Cade, Dean, Davis Jones, and Brennecke)
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, Melbourne, Australia (Dr Brennecke)
| |
Collapse
|
155
|
Giorgione V, Di Fabrizio C, Giallongo E, Khalil A, O'Driscoll J, Whitley G, Kennedy G, Murdoch CE, Thilaganathan B. Angiogenic markers and maternal echocardiographic indices in women with hypertensive disorders of pregnancy. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2024; 63:206-213. [PMID: 37675647 DOI: 10.1002/uog.27474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/11/2023] [Accepted: 08/24/2023] [Indexed: 09/08/2023]
Abstract
OBJECTIVE The maternal cardiovascular system of women with hypertensive disorders of pregnancy (HDP) can be impaired, with higher rates of left ventricular (LV) remodeling and diastolic dysfunction compared to those with normotensive pregnancy. The primary objective of this prospective study was to correlate cardiac indices obtained by transthoracic echocardiography (TTE) and circulating angiogenic markers, such as soluble fms-like tyrosine kinase-1 (sFlt-1) and placental growth factor (PlGF). METHODS In this study, 95 women with a pregnancy complicated by HDP and a group of 25 with an uncomplicated pregnancy at term underwent TTE and blood tests to measure sFlt-1 and PlGF during the peripartum period (before delivery or within a week of giving birth). Spearman's rank correlation was used to derive correlation coefficients between biomarkers and cardiac indices in the HDP and control populations. RESULTS The HDP group included 61 (64.2%) pre-eclamptic patients and, among them, 42 (68.9%) delivered before 37 weeks' gestation. Twelve women with HDP (12.6%) underwent blood sampling and TTE after delivery, and, as they showed significantly lower levels of angiogenic markers, they were excluded from the analysis. There was a correlation between sFlt-1 and LV mass index (LVMI) (r = 0.246; P = 0.026) and early diastolic mitral inflow velocity (E) and early diastolic mitral annular velocity (e') ratio (r = 0.272; P = 0.014) in the HDP group (n = 83), while in the controls, sFlt-1 showed a correlation with relative wall thickness (r = 0.409; P = 0.043), lateral e' (r = -0.562; P = 0.004) and E/e' ratio (r = 0.417; P = 0.042). PlGF correlated with LVMI (r = -0.238; P = 0.031) in HDP patients and with lateral e' (r = 0.466; P = 0.022) in controls. sFlt-1/PlGF ratio correlated with lateral e' (r = -0.568; P = 0.004) and E/e' ratio (r = 0.428; P = 0.037) in controls and with LVMI (r = 0.252; P = 0.022) and E/e' ratio (r = 0.269; P = 0.014) in HDP. CONCLUSIONS Although the current data are not able to infer causality, they confirm the intimate relationship between the maternal cardiovascular system and angiogenic markers that are used both to diagnose and indicate the severity of HDP. © 2023 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- V Giorgione
- Vascular Biology Research Centre, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust, University of London, London, UK
| | - C Di Fabrizio
- Vascular Biology Research Centre, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - E Giallongo
- Intensive Care National Audit & Research Centre, London, UK
| | - A Khalil
- Vascular Biology Research Centre, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust, University of London, London, UK
| | - J O'Driscoll
- Department of Cardiology, St George's University Hospitals NHS Foundation Trust, London, UK
- School of Psychology and Life Sciences, Canterbury Christ Church University, Kent, UK
| | - G Whitley
- Vascular Biology Research Centre, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - G Kennedy
- Immunoassay Biomarker Core Laboratory, School of Medicine, University of Dundee, Dundee, UK
| | - C E Murdoch
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - B Thilaganathan
- Vascular Biology Research Centre, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust, University of London, London, UK
| |
Collapse
|
156
|
Pluta K, Januszewski M, Ziuzia-Januszewska L, Kudan M, Suchocka M, Kuśmierczuk K, Issat T, Jakimiuk AJ. sFlT-1/PlGF ratio as a predictor of preeclampsia in COVID-19 pregnant patients. BMC Pregnancy Childbirth 2024; 24:94. [PMID: 38297191 PMCID: PMC10829318 DOI: 10.1186/s12884-024-06263-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 01/07/2024] [Indexed: 02/02/2024] Open
Abstract
The association between SARS-CoV-2 infection in pregnancy and preeclampsia is widely debated in numerous studies. The aim of our study was to investigate whether an increased sFlt-1/PlGF ratio is a good marker of preeclampsia in pregnant patients with COVID-19 infection. This single centre prospective study was conducted in the Department of Obstetrics and Gynaecology, at the Central Clinical Hospital of the Ministry of the Interior and Administration in Warsaw. The study group consisted of 68 COVID-19 pregnant patients and 57 SARS-CoV-2 negative pregnant controls. Serum sFlt-1/PlGF ratio was assessed. The two groups did not differ in terms of the frequency of IVF, nulliparity, history of hypertension, pre-gestational diabetes and chronic kidney disease. The primary outcome was the diagnosis of preeclampsia. Preeclampsia was diagnosed in 10 patients in both groups. The sFlt-1/PlGF ratio higher than 38, considered highly suggestive of developing preeclampsia, was found in 20 patients in the COVID-19 group and 15 patients in the control group. The odds of developing preeclampsia in patients with sFlt-1/PlGF ratio > 38 was approximately 4-fold higher in COVID-19 group and 11-fold higher in controls. Sflt-1/PlGF ratio does not differ significantly between the SARS-CoV-2-positive and SARS-COV-2-negative pregnant patients. The sFlt-1/PlGF ratio > 38 is associated with higher odds of the diagnosis of preeclampsia in both of these groups, and therefore may serve as its marker regardless of COVID-19 infection status.
Collapse
Affiliation(s)
- Kamil Pluta
- Department of Obstetrics and Gynecology, National Medical Institute of the Ministry of Interior and Administration, Warsaw, Poland
| | - Marcin Januszewski
- Department of Obstetrics and Gynecology, National Medical Institute of the Ministry of Interior and Administration, Warsaw, Poland
| | - Laura Ziuzia-Januszewska
- Department of Otolaryngology, National Medical Institute of the Ministry of Interior and Administration, Warsaw, Poland
| | - Michał Kudan
- Department of Obstetrics and Gynecology, National Medical Institute of the Ministry of Interior and Administration, Warsaw, Poland
| | - Maria Suchocka
- Department of Obstetrics and Gynecology, National Medical Institute of the Ministry of Interior and Administration, Warsaw, Poland
| | - Kinga Kuśmierczuk
- Department of Obstetrics and Gynecology, National Medical Institute of the Ministry of Interior and Administration, Warsaw, Poland
| | - Tadeusz Issat
- Department of Obstetrics and Gynecology, Institute of Mother and Child, Warsaw, Poland
| | - Artur J Jakimiuk
- Department of Obstetrics and Gynecology, National Medical Institute of the Ministry of Interior and Administration, Warsaw, Poland.
- Center for Reproductive Health, Institute of Mother and Child, Warsaw, Poland.
| |
Collapse
|
157
|
Medegan Fagla B, Buhimschi IA. Protein Misfolding in Pregnancy: Current Insights, Potential Mechanisms, and Implications for the Pathogenesis of Preeclampsia. Molecules 2024; 29:610. [PMID: 38338354 PMCID: PMC10856193 DOI: 10.3390/molecules29030610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
Protein misfolding disorders are a group of diseases characterized by supra-physiologic accumulation and aggregation of pathogenic proteoforms resulting from improper protein folding and/or insufficiency in clearance mechanisms. Although these processes have been historically linked to neurodegenerative disorders, such as Alzheimer's disease, evidence linking protein misfolding to other pathologies continues to emerge. Indeed, the deposition of toxic protein aggregates in the form of oligomers or large amyloid fibrils has been linked to type 2 diabetes, various types of cancer, and, in more recent years, to preeclampsia, a life-threatening pregnancy-specific disorder. While extensive physiological mechanisms are in place to maintain proteostasis, processes, such as aging, genetic factors, or environmental stress in the form of hypoxia, nutrient deprivation or xenobiotic exposures can induce failure in these systems. As such, pregnancy, a natural physical state that already places the maternal body under significant physiological stress, creates an environment with a lower threshold for aberrant aggregation. In this review, we set out to discuss current evidence of protein misfolding in pregnancy and potential mechanisms supporting a key role for this process in preeclampsia pathogenesis. Improving our understanding of this emerging pathophysiological process in preeclampsia can lead to vital discoveries that can be harnessed to create better diagnoses and treatment modalities for the disorder.
Collapse
Affiliation(s)
| | - Irina Alexandra Buhimschi
- Department of Obstetrics and Gynecology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA;
| |
Collapse
|
158
|
Zhu Y, Wu B, Gao X, Guo Y, Peng X, Wang L, Tian X, Wang J, Yan W, Zhang H, Qu J. Fluorescence lifetime imaging microscopy for early diagnosis and severity prediction of preeclampsia with Nile Blue probe. Biosens Bioelectron 2024; 244:115809. [PMID: 37931439 DOI: 10.1016/j.bios.2023.115809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/22/2023] [Accepted: 10/30/2023] [Indexed: 11/08/2023]
Abstract
Preeclampsia (PE) is a common pregnancy complication and the leading cause of maternal and perinatal mortality. Unfortunately, the early diagnostic methods for PE are still rare. Fluorescence lifetime imaging (FLIM) technology has proven to be applicable for diagnosis of various diseases. Here, we explore the possibility of the FLIM technique for PE early diagnosis and severity prediction with Nile Blue probe as biosensor. 23 placental slices and 162 third-trimester-collected maternal peripheral blood serum samples were stained with Nile blue and imaged by FLIM system. Fluorescence lifetimes of the probe increased significantly as the disease worsened (p < 0.0001). Characterization of the probe showed an increasing tendency in lifetimes under lower polarity conditions and revealed that the reason for the lifetime differences in serum sample was polarity changes caused by abnormal lipid metabolism in serum. For early diagnosis, we investigated 42 12th-week-collected chronic hypertension (CH) serum samples and successfully distinguished PE patients from pregnant women. With the functions of measuring fluorescence lifetime and detecting polarity changes caused by an abnormal lipid microenvironment in maternal peripheral blood, FLIM technology, together with Nile Blue probe, presents a feasible and advantageous approach for PE early noninvasive diagnosis and severity prediction.
Collapse
Affiliation(s)
- Yinru Zhu
- College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, China
| | - Benqing Wu
- University of Chinese Academy of Sciences Shenzhen Hospital, Guangming District, Shenzhen, 518106, China
| | - Xinwei Gao
- College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, China
| | - Yong Guo
- College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, China
| | - Xiao Peng
- College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, China
| | - Luwei Wang
- College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, China
| | - Xuewu Tian
- University of Chinese Academy of Sciences Shenzhen Hospital, Guangming District, Shenzhen, 518106, China
| | - Jian Wang
- College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, China
| | - Wei Yan
- College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, China.
| | - Hui Zhang
- University of Chinese Academy of Sciences Shenzhen Hospital, Guangming District, Shenzhen, 518106, China.
| | - Junle Qu
- College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, China
| |
Collapse
|
159
|
Bacmeister L, Goßling A, Buellesbach A, Birukov A, Myers JE, Thomas ST, Lee S, Andersen MS, Jorgensen JS, Diemert A, Blois SM, Arck PC, Hecher K, Herse F, Blankenberg S, Dechend R, Westermann D, Zeller T. High-Sensitivity Cardiac Troponin I Enhances Preeclampsia Prediction Beyond Maternal Factors and the sFlt-1/PlGF Ratio. Circulation 2024; 149:95-106. [PMID: 37982257 DOI: 10.1161/circulationaha.123.066199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/13/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Preeclampsia shares numerous risk factors with cardiovascular diseases. Here, we aimed to assess the potential utility of high-sensitivity cardiac troponin I (hs-cTnI) values during pregnancy in predicting preeclampsia occurrence. METHODS This study measured hs-cTnI levels in 3721 blood samples of 2245 pregnant women from 4 international, prospective cohorts. Three analytical approaches were used: (1) a cross-sectional analysis of all women using a single blood sample, (2) a longitudinal analysis of hs-cTnI trajectories in women with multiple samples, and (3) analyses of prediction models incorporating hs-cTnI, maternal factors, and the sFlt-1 (soluble fms-like tyrosine kinase 1)/PlGF (placental growth factor) ratio. RESULTS Women with hs-cTnI levels in the upper quarter had higher odds ratios for preeclampsia occurrence compared with women with levels in the lower quarter. Associations were driven by preterm preeclampsia (odds ratio, 5.78 [95% CI, 2.73-12.26]) and remained significant when using hs-cTnI as a continuous variable adjusted for confounders. Between-trimester hs-cTnI trajectories were independent of subsequent preeclampsia occurrence. A prediction model incorporating a practical hs-cTnI level of detection cutoff (≥1.9 pg/mL) alongside maternal factors provided comparable performance with the sFlt-1/PlGF ratio. A comprehensive model including sFlt-1/PlGF, maternal factors, and hs-cTnI provided added value (cross-validated area under the receiver operator characteristic, 0.78 [95% CI, 0.73-0.82]) above the sFlt-1/PlGF ratio alone (cross-validated area under the receiver operator characteristic, 0.70 [95% CI, 0.65-0.76]; P=0.027). As assessed by likelihood ratio tests, the addition of hs-cTnI to each prediction model significantly improved the respective prediction model not incorporating hs-cTnI, particularly for preterm preeclampsia. Net reclassification improvement analyses indicated that incorporating hs-cTnI improved risk prediction predominantly by correctly reclassifying women with subsequent preeclampsia occurrence. CONCLUSIONS These exploratory findings uncover a potential role for hs-cTnI as a complementary biomarker in the prediction of preeclampsia. After validation in prospective studies, hs-cTnI, alongside maternal factors, may either be considered as a substitute for angiogenic biomarkers in health care systems where they are sparce or unavailable, or as an enhancement to established prediction models using angiogenic markers.
Collapse
Affiliation(s)
- Lucas Bacmeister
- Clinic for Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany (L.B., A. Buellesbach, D.W.)
| | - Alina Goßling
- Department of Cardiology (A.G., S.B., T.Z.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Annette Buellesbach
- Clinic for Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany (L.B., A. Buellesbach, D.W.)
| | - Anna Birukov
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (A. Birukov)
- Department of Molecular Epidemiology, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal (A. Birukov)
| | - Jenny E Myers
- Maternal & Fetal Health Research Centre, Faculty Biology, Medicine & Health, University of Manchester, United Kingdom (J.E.M., S.T.T., S.L.)
| | - Susan T Thomas
- Maternal & Fetal Health Research Centre, Faculty Biology, Medicine & Health, University of Manchester, United Kingdom (J.E.M., S.T.T., S.L.)
| | - Stacy Lee
- Maternal & Fetal Health Research Centre, Faculty Biology, Medicine & Health, University of Manchester, United Kingdom (J.E.M., S.T.T., S.L.)
| | - Marianne S Andersen
- Department of Endocrinology, Odense University Hospital (M.S.A.), University of Southern Denmark, Odense
| | - Jan S Jorgensen
- Institute for Clinical Research, Faculty of Health Sciences (J.S.J.), University of Southern Denmark, Odense
| | - Anke Diemert
- Department of Obstetrics and Fetal Medicine (A.D., S.M.B., P.C.A., K.H.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra M Blois
- Department of Obstetrics and Fetal Medicine (A.D., S.M.B., P.C.A., K.H.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petra C Arck
- Department of Obstetrics and Fetal Medicine (A.D., S.M.B., P.C.A., K.H.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kurt Hecher
- Department of Obstetrics and Fetal Medicine (A.D., S.M.B., P.C.A., K.H.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Herse
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (F.H., R.D.)
| | - Stefan Blankenberg
- Department of Cardiology (A.G., S.B., T.Z.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- University Center for Cardiovascular Research (S.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research, partner site Hamburg/Kiel/Lübeck, Hamburg (S.B., T.Z.)
| | - Ralf Dechend
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (F.H., R.D.)
- HELIOS Clinic Berlin-Buch, Department of Cardiology and Nephrology, Berlin, Germany (R.D.)
| | - Dirk Westermann
- Clinic for Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany (L.B., A. Buellesbach, D.W.)
| | - Tanja Zeller
- Department of Cardiology (A.G., S.B., T.Z.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research, partner site Hamburg/Kiel/Lübeck, Hamburg (S.B., T.Z.)
| |
Collapse
|
160
|
Anto EO, Boadu WIO, Hughes C, Korsah EE, Frimpong J, Ansah E, Tawiah A, Tamakloe VCKT, Acheampong E, Asamoah EA, Opoku S, Aboagye E, Albright EM, Adua E, Afrifa-Yamoah E, Anto AO, Annani-Akollor ME, Wiafe YA, Obirikorang C. Angiogenic growth factors, oxidative stress and haematobiochemical measures as predictors of preeclampsia with and without foetal growth restriction: A case-control study in a Ghanaian population. Placenta 2024; 145:130-138. [PMID: 38134545 DOI: 10.1016/j.placenta.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 11/27/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023]
Abstract
INTRODUCTION This study compares the angiogenic growth mediators (AGMs), oxidative stress (OS) and haematobiochemical profile as well as foeto-maternal outcomes of preeclampsia (PE) with and without foetal growth restriction (FGR) and the discriminative potential of these markers for identifying these conditions. METHODS This hospital-based case-control study recruited a total of 209 women including 109 PE women without FGR and 48 PE women with FGR as cases whereas 52 normotensive pregnant women were recruited as controls. OS and AGMs and haematobiochemical markers were measured for all participants. RESULTS The rates of foetal complications including intrauterine foetal death and foetal distress were more common in PE with FGR than PE without FGR (p < 0.05) but maternal complications were comparable across these groups (p > 0.05). Of the haematobiochemical markers, placental growth factors (PIGF), PIGF/8-Isoprostane, sFlt-1/PIGF (AUC = 0.87, p < 0.001), soluble FMS-tyrosine kinase receptor-1 (sFlt-1) (AUC = 0.85, p < 0.001), total antioxidant capacity, 8-isoprostane (AUC = 0.83, p < 0.001) and lactate dehydrogenase (AUC = 0.70, p < 0.001) were more associated and showed at least an acceptable discrimination for PE with FGR against PE only. DISCUSSION The occurrence of FGR in PE patients does not necessarily indicate a severe maternal presentation of the condition but a tendency for adverse foetal outcomes. Cumulative assessment of OS and AGMs may provide diagnostic usefulness for distinguishing PE with and without FGR.
Collapse
Affiliation(s)
- Enoch Odame Anto
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana; School of Medical and Health Sciences, Edith Cowan University, Joondalup Drive, Perth, Australia; Centre for Precision Health, ECU Strategic Research Centre, Edith Cowan University, Perth, Australia.
| | - Wina Ivy Ofori Boadu
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Charity Hughes
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Emmanuel Ekow Korsah
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Joseph Frimpong
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Ezekiel Ansah
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | | | | | - Emmanuel Acheampong
- Centre for Precision Health, ECU Strategic Research Centre, Edith Cowan University, Perth, Australia
| | - Evans Adu Asamoah
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Health Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Stephen Opoku
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Health Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Elizabeth Aboagye
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Etwi-Mensah Albright
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Eric Adua
- Rural Clinical School, Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | | | - Agartha Odame Anto
- Department of Obstetrics and Gynaecology, Ho Teaching Hospital, Ho, Ghana
| | - Max Efui Annani-Akollor
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Health Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Yaw Amo Wiafe
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Christian Obirikorang
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Health Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
161
|
Sentilhes L, Schmitz T, Arthuis C, Barjat T, Berveiller P, Camilleri C, Froeliger A, Garabedian C, Guerby P, Korb D, Lecarpentier E, Mattuizzi A, Sibiude J, Sénat MV, Tsatsaris V. [Preeclampsia: Guidelines for clinical practice from the French College of Obstetricians and Gynecologists]. GYNECOLOGIE, OBSTETRIQUE, FERTILITE & SENOLOGIE 2024; 52:3-44. [PMID: 37891152 DOI: 10.1016/j.gofs.2023.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
OBJECTIVE To identify strategies to reduce maternal and neonatal morbidity related to preeclampsia. MATERIAL AND METHODS The quality of evidence of the literature was assessed following the GRADE® method with questions formulated in the PICO format (Patients, Intervention, Comparison, Outcome) and outcomes defined a priori and classified according to their importance. An extensive bibliographic search was performed on PubMed, Cochrane, EMBASE and Google Scholar databases. The quality of the evidence was assessed (high, moderate, low, very low) and recommendations were formulated as a (i) strong, (ii) weak or (iii) no recommendation. The recommendations were reviewed in two rounds with external reviewers (Delphi survey) to select the consensus recommendations. RESULTS Preeclampsia is defined by the association of gestational hypertension (systolic blood pressure≥140mmHg and/or diastolic blood pressure≥90mmHg) and proteinuria≥0.3g/24h or a Proteinuria/Creatininuria ratio≥30mg/mmol occurring after 20 weeks of gestation. Data from the literature do not show any benefit in terms of maternal or perinatal health from implementing a broader definition of preeclampsia. Of the 31 questions, there was agreement between the working group and the external reviewers on 31 (100%). In general population, physical activity during pregnancy should be encouraged to reduce the risk of preeclampsia (Strong recommendation, Quality of the evidence low) but an early screening based on algorithms (Weak recommendation, Quality of the evidence low) or aspirin administration (Weak recommendation, Quality of the evidence very low) is not recommended to reduce maternal and neonatal morbidity related to preeclampsia. In women with preexisting diabetes or hypertension or renal disease, or multiple pregnancy, the level of evidence is insufficient to determine whether aspirin administration during pregnancy is useful to reduce maternal and perinatal morbidity (No recommendation, Quality of the evidence low). In women with a history of vasculo-placental disease, low dose of aspirin (Strong recommendation, Quality of the evidence moderate) at a dosage of 100-160mg per day (Weak recommendation, Quality of the evidence low), ideally before 16 weeks of gestation and not after 20 weeks of gestation (Strong recommendation, Quality of the evidence low) until 36 weeks of gestation (Weak recommendation, Quality of the evidence very low) is recommended. In a high-risk population, additional administration of low molecular weight heparin is not recommended (Weak recommendation, Quality of the evidence moderate). In case of preeclampsia (Weak recommendation, Quality of the evidence low) or suspicion of preeclampsia (Weak recommendation, Quality of the evidence moderate, the assessment of PlGF concentration or sFLT-1/PlGF ratio is not routinely recommended) in the only goal to reduce maternal or perinatal morbidity. In women with non-severe preeclampsia antihypertensive agent should be administered orally when the systolic blood pressure is measured between 140 and 159mmHg or diastolic blood pressure is measured between 90 and 109mmHg (Weak recommendation, Quality of the evidence low). In women with non-severe preeclampsia, delivery between 34 and 36+6 weeks of gestation reduces severe maternal hypertension but increases the incidence of moderate prematurity. Taking into account the benefit/risk balance for the mother and the child, it is recommended not to systematically induce birth in women with non-severe preeclampsia between 34 and 36+6 weeks of gestation (Strong recommendation, Quality of evidence high). In women with non-severe preeclampsia diagnosed between 37+0 and 41 weeks of gestation, it is recommended to induce birth to reduce maternal morbidity (Strong recommendation, Low quality of evidence), and to perform a trial of labor in the absence of contraindication (Strong recommendation, Very low quality of evidence). In women with a history of preeclampsia, screening maternal thrombophilia is not recommended (Strong recommendation, Quality of the evidence moderate). Because women with a history of a preeclampsia have an increased lifelong risk of chronic hypertension and cardiovascular complications, they should be informed of the need for medical follow-up to monitor blood pressure and to manage other possible cardiovascular risk factors (Strong recommendation, Quality of the evidence moderate). CONCLUSION The purpose of these recommendations was to reassess the definition of preeclampsia, and to determine the strategies to reduce maternal and perinatal morbidity related to preeclampsia, during pregnancy but also after childbirth. They aim to help health professionals in their daily clinical practice to inform or care for patients who have had or have preeclampsia. Synthetic information documents are also offered for professionals and patients.
Collapse
Affiliation(s)
- Loïc Sentilhes
- Service de gynécologie-obstétrique, centre hospitalier universitaire de Bordeaux, Bordeaux, France.
| | - Thomas Schmitz
- Service de gynécologie-obstétrique, hôpital Robert-Debré, AP-HP, Paris, France
| | - Chloé Arthuis
- Service d'obstétrique et de médecine fœtale, Elsan Santé Atlantique, 44819 Saint-Herblain, France
| | - Tiphaine Barjat
- Service de gynécologie-obstétrique, centre hospitalier universitaire de Saint-Etienne, Saint-Etienne, France
| | - Paul Berveiller
- Service de gynécologie-obstétrique, centre hospitalier intercommunal de Poissy St-Germain, Poissy, France
| | - Céline Camilleri
- Association grossesse santé contre la pré-éclampsie, Paris, France
| | - Alizée Froeliger
- Service de gynécologie-obstétrique, centre hospitalier universitaire de Bordeaux, Bordeaux, France
| | - Charles Garabedian
- Service de gynécologie-obstétrique, University Lille, ULR 2694-METRICS, CHU de Lille, 59000 Lille, France
| | - Paul Guerby
- Service de gynécologie-obstétrique, centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Diane Korb
- Service de gynécologie-obstétrique, hôpital Robert-Debré, AP-HP, Paris, France
| | - Edouard Lecarpentier
- Service de gynécologie-obstétrique, centre hospitalier intercommunal de Créteil, Créteil, France
| | - Aurélien Mattuizzi
- Service de gynécologie-obstétrique, centre hospitalier universitaire de Bordeaux, Bordeaux, France
| | - Jeanne Sibiude
- Service de gynécologie-obstétrique, hôpital Louis-Mourier, AP-HP, Colombes, France
| | - Marie-Victoire Sénat
- Service de gynécologie-obstétrique, hôpital Bicêtre, AP-HP, Le Kremlin-Bicêtre, France
| | - Vassilis Tsatsaris
- Maternité Port-Royal, hôpital Cochin, GHU Centre Paris cité, AP-HP, FHU PREMA, Paris, France
| |
Collapse
|
162
|
Singh Thakur A, Tayade S, Patel D, Gupta A, Batra N. Unraveling the Predictive Power: Placenta Growth Factor and Pregnancy-Associated Plasma Protein A in Pre-eclampsia. Cureus 2024; 16:e52752. [PMID: 38389635 PMCID: PMC10881904 DOI: 10.7759/cureus.52752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
This review provides a comprehensive exploration of the roles of placenta growth factor (PlGF) and pregnancy-associated plasma protein A (PAPP-A) in the context of pre-eclampsia, a pregnancy-related hypertensive disorder with significant implications for maternal and fetal health. The background elucidates the clinical significance of pre-eclampsia, highlighting its prevalence and impact. The review delves into the biological importance of PlGF and PAPP-A, emphasizing their critical roles in normal placental development and their dysregulation in pre-eclampsia. Notably, altered levels of these biomarkers emerge as potential diagnostic indicators, offering insights into the pathophysiology of the disorder. The exploration of pathophysiological mechanisms, including angiogenic imbalance and placental dysfunction, provides a nuanced understanding of pre-eclampsia's molecular landscape. The therapeutic implications of targeting PlGF and PAPP-A open avenues for future research, aiming at effective intervention strategies. The conclusion summarizes key findings, outlines implications for future research, and underscores the crucial role of PlGF and PAPP-A in understanding and managing pre-eclampsia, with the ultimate goal of improving outcomes for both mothers and infants.
Collapse
Affiliation(s)
- Aditi Singh Thakur
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Wardha, IND
| | - Surekha Tayade
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Wardha, IND
| | - Drashti Patel
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Wardha, IND
| | - Aishwarya Gupta
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Wardha, IND
| | - Nitish Batra
- Medicine, Jawaharlal Nehru Medical College, Wardha, IND
| |
Collapse
|
163
|
Sokratous N, Wright A, Syngelaki A, Kakouri E, Laich A, Nicolaides KH. Screening for pre-eclampsia by maternal serum glycosylated fibronectin and angiogenic markers at 36 weeks' gestation. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2024; 63:88-97. [PMID: 37724582 DOI: 10.1002/uog.27481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/21/2023]
Abstract
OBJECTIVES First, to examine the predictive performance of maternal serum glycosylated fibronectin (GlyFn) at 35 + 0 to 36 + 6 weeks' gestation in screening for delivery with pre-eclampsia (PE) and delivery with gestational hypertension (GH) at ≥ 37 weeks' gestation, both within 3 weeks and at any time after the examination. Second, to compare the predictive performance for delivery with PE and delivery with GH of various combinations of biomarkers, including GlyFn, mean arterial pressure (MAP), uterine artery pulsatility index (UtA-PI), serum placental growth factor (PlGF) and soluble fms-like tyrosine kinase-1 (sFlt-1). Third, to compare the predictive performance for delivery with PE and delivery with GH by serum PlGF concentration, sFlt-1/PlGF concentration ratio and the competing-risks model with different combinations of biomarkers as above. Fourth, to compare the predictive performance of screening at 11 + 0 to 13 + 6 weeks vs 35 + 0 to 36 + 6 weeks for delivery with PE and delivery with GH at ≥ 37 weeks' gestation. METHODS This was a case-control study in which maternal serum GlyFn was measured in stored samples from a non-intervention screening study in singleton pregnancies at 35 + 0 to 36 + 6 weeks' gestation using a point-of-care device. We used samples from women who delivered at ≥ 37 weeks' gestation, including 100 who developed PE, 100 who developed GH and 600 controls who did not develop PE or GH. In all cases, MAP, UtA-PI, PlGF and sFlt-1 were measured during the routine visit at 35 + 0 to 36 + 6 weeks. We used samples from patients that had been examined previously at 11 + 0 to 13 + 6 weeks' gestation. Levels of GlyFn were transformed to multiples of the expected median (MoM) values after adjusting for maternal demographic characteristics and elements from the medical history. Similarly, the measured values of MAP, UtA-PI, PlGF and sFlt-1 were converted to MoM. The competing-risks model was used to combine the prior distribution of the gestational age at delivery with PE, obtained from maternal risk factors, with various combinations of biomarker MoM values to derive the patient-specific risks of delivery with PE. The performance of screening of different strategies was estimated by examining the detection rate (DR) at a 10% fixed false-positive rate (FPR) and McNemar's test was used to compare the DRs between the different methods of screening. RESULTS The DR, at 10% FPR, of screening by the triple test (maternal risk factors plus MAP, PlGF and sFlt-1) was 83.7% (95% CI, 70.3-92.7%) for delivery with PE within 3 weeks of screening and 80.0% (95% CI, 70.8-87.3%) for delivery with PE at any time after screening, and this performance was not improved by the addition of GlyFn. The performance of screening by a combination of maternal risk factors, MAP, PlGF and GlyFn was similar to that of the triple test, both for delivery with PE within 3 weeks and at any time after screening. The performance of screening by a combination of maternal risk factors, MAP, UtA-PI and GlyFn was similar to that of the triple test, and they were both superior to screening by low PlGF concentration (PE within 3 weeks: DR, 65.3% (95% CI, 50.4-78.3%); PE at any time: DR, 56.0% (95% CI, 45.7-65.9%)) or high sFlt-1/PlGF concentration ratio (PE within 3 weeks: DR, 73.5% (95% CI, 58.9-85.1%); PE at any time: DR, 63.0% (95% CI, 52.8-72.4%)). The predictive performance of screening at 35 + 0 to 36 + 6 weeks' gestation for delivery with PE and delivery with GH at ≥ 37 weeks' gestation was by far superior to screening at 11 + 0 to 13 + 6 weeks. CONCLUSION GlyFn is a potentially useful biomarker in third-trimester screening for term PE and term GH, but the findings of this case-control study need to be validated by prospective screening studies. © 2023 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- N Sokratous
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - A Wright
- Institute of Health Research, University of Exeter, Exeter, UK
| | - A Syngelaki
- Fetal Medicine Research Institute, King's College Hospital, London, UK
- Institute of Women and Children's Health, School of Life Course and Population Sciences, King's College London, London, UK
| | - E Kakouri
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - A Laich
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - K H Nicolaides
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| |
Collapse
|
164
|
Nobleza COS. Neurologic complications in the obstetric patient. THE BRAIN OF THE CRITICALLY ILL PREGNANT WOMAN 2024:3-40. [DOI: 10.1016/b978-0-443-15205-4.00005-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
165
|
Morey R, Poling L, Srinivasan S, Martinez-King C, Anyikam A, Zhang-Rutledge K, To C, Hakim A, Mochizuki M, Verma K, Mason A, Tran V, Meads M, Lamale-Smith L, Roeder H, Horii M, Ramos GA, DeHoff P, Parast MM, Pantham P, Laurent LC. Discovery and verification of extracellular microRNA biomarkers for diagnostic and prognostic assessment of preeclampsia at triage. SCIENCE ADVANCES 2023; 9:eadg7545. [PMID: 38117879 PMCID: PMC10732528 DOI: 10.1126/sciadv.adg7545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 11/17/2023] [Indexed: 12/22/2023]
Abstract
We report on the identification of extracellular miRNA (ex-miRNA) biomarkers for early diagnosis and prognosis of preeclampsia (PE). Small RNA sequencing of maternal serum prospectively collected from participants undergoing evaluation for suspected PE revealed distinct patterns of ex-miRNA expression among different categories of hypertensive disorders in pregnancy. Applying an iterative machine learning method identified three bivariate miRNA biomarkers (miR-522-3p/miR-4732-5p, miR-516a-5p/miR-144-3p, and miR-27b-3p/let-7b-5p) that, when applied serially, distinguished between PE cases of different severity and differentiated cases from controls with a sensitivity of 93%, specificity of 79%, positive predictive value (PPV) of 55%, and negative predictive value (NPV) of 89%. In a small independent validation cohort, these ex-miRNA biomarkers had a sensitivity of 91% and specificity of 57%. Combining these ex-miRNA biomarkers with the established sFlt1:PlGF protein biomarker ratio performed better than either set of biomarkers alone (sensitivity of 89.4%, specificity of 91.3%, PPV of 95.5%, and NPV of 80.8%).
Collapse
Affiliation(s)
- Robert Morey
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Lara Poling
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Srimeenakshi Srinivasan
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Carolina Martinez-King
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Adanna Anyikam
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Kathy Zhang-Rutledge
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Cuong To
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Abbas Hakim
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Marina Mochizuki
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Kajal Verma
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Antoinette Mason
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Vy Tran
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Morgan Meads
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Leah Lamale-Smith
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Hilary Roeder
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Mariko Horii
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Gladys A. Ramos
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Peter DeHoff
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Mana M. Parast
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Priyadarshini Pantham
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Louise C. Laurent
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
166
|
Zhu C, Xu CJ, Wu JN, Zhao W, Hu YL, Yao Y, Ren YY. Association between abnormal uterine artery pulsatility index and the risk of fetal congenital heart defects: a hospital-based cohort study. Sci Rep 2023; 13:22924. [PMID: 38129577 PMCID: PMC10739791 DOI: 10.1038/s41598-023-50167-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
To explore the associations between high uterine artery pulsatility index (UtA-PI) values and congenital heart disease (CHD) risk and whether they differed between singleton and multiple pregnancies. This hospital-based cohort study involving 52,047 pregnant women who underwent prenatal examinations from 2012 to 2016. Infants born to the included pregnant women were followed until 42 days after birth to identify those with CHDs. Generalized estimating equations were used to estimate the associations of high right UtA-PI (> 95th percentile) values with maternal preeclampsia and fetal CHDs. Logistic regression analyses were conducted using path analysis models to quantify the effect of high right UtA-PI values on fetal CHD risk. A total of 42,552 women and 43,470 infants (147 with CHDs) were included. Preeclampsia risk was associated with a high right UtA-PI in singleton-pregnant women (adjusted PR, 3.01; 95% CI 2.57-3.52). CHD risk was marginally associated with a high right UtA-PI in singleton-pregnant women (adjusted PR, 2.26, 95% CI 1.03-4.95). Considering only two factors, 96.0% of the fetal CHD risk was mediated by preeclampsia in singleton-pregnant women, while 93.8% of the risk was related to a high right UtA-PI in multiple-pregnant women. A high right UtA-PI was marginally associated with an increased fetal CHD risk in singleton-pregnant women and might play an important role in multiple-pregnant women. Further studies are warranted to confirm these findings given the high loss to follow-up rate.
Collapse
Affiliation(s)
- Chen Zhu
- Department of Ultrasound, Obstetrics and Gynecology Hospital of Fudan University, No. 588 Fangxie Road, Shanghai, China
| | - Cheng-Jie Xu
- Department of Information Technology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jiang-Nan Wu
- Department of Clinical Epidemiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Wei Zhao
- Department of Ultrasound, Obstetrics and Gynecology Hospital of Fudan University, No. 588 Fangxie Road, Shanghai, China
| | - Yan-Lai Hu
- Department of Ultrasound, Obstetrics and Gynecology Hospital of Fudan University, No. 588 Fangxie Road, Shanghai, China
| | - Ying Yao
- Department of Ultrasound, Obstetrics and Gynecology Hospital of Fudan University, No. 588 Fangxie Road, Shanghai, China
| | - Yun-Yun Ren
- Department of Ultrasound, Obstetrics and Gynecology Hospital of Fudan University, No. 588 Fangxie Road, Shanghai, China.
| |
Collapse
|
167
|
Štritof PG, Zanze L, Zekan P, Horvat G, Blagaic V. Mirror syndrome associated with fetal cardiomyopathy. Arch Clin Cases 2023; 10:191-195. [PMID: 38098693 PMCID: PMC10719984 DOI: 10.22551/2023.41.1004.10273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023] Open
Abstract
Mirror syndrome is a rare condition of generalized maternal oedema caused by fetal hydrops. A 37-year-old patient was admitted to our hospital because of suspected mirror syndrome caused by fetal cardiomyopathy. At 26th week of gestation patient developed bilateral pulmonary oedema as her condition rapidly deteriorated. Consequently, preterm labor was induced, percutaneous evacuation of fetal ascites was performed, and the patient finally vaginally delivered stillborn fetus. Although the initial postpartum period was severely complicated by hemorrhage, the condition of the patient significantly improved later, and she was discharged seven days after delivery. We believe this case is worth presenting due to its rarity and significant perinatal and obstetric challenges in treatment of those patients. Furthermore, preimplantation genetic testing could be performed to prevent at least some of the cases.
Collapse
Affiliation(s)
- Petra Glad Štritof
- Department of Obstetrics and Gynecology, University Hospital “Sveti Duh“, Zagreb, Croatia
| | - Lucija Zanze
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Paulo Zekan
- Department of Obstetrics and Gynecology, University Hospital “Sveti Duh“, Zagreb, Croatia
| | | | - Vladimir Blagaic
- Department of Obstetrics and Gynecology, University Hospital “Sveti Duh“, Zagreb, Croatia
| |
Collapse
|
168
|
Ashley RL, Trigo EM, Ervin JM. Placental insufficiency and heavier placentas in sheep after suppressing CXCL12/CXCR4 signaling during implantation†. Biol Reprod 2023; 109:982-993. [PMID: 37724932 PMCID: PMC10724462 DOI: 10.1093/biolre/ioad122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/28/2023] [Accepted: 09/15/2023] [Indexed: 09/21/2023] Open
Abstract
During implantation, trophoblast cell invasion and differentiation is predominantly important to achieving proper placental formation and embryonic development. The chemokine, C-X-C motif chemokine ligand 12 (CXCL12) working through its receptor C-X-C motif chemokine receptor 4 (CXCR4) is implicated in implantation and placentation but precise roles of this axis are unclear. Suppressing CXCL12/CXCR4 signaling at the fetal-maternal interface in sheep reduces trophoblast invasion, disrupts uterine remodeling, and diminishes placental vascularization. We hypothesize these negative impacts during implantation will manifest as compromised fetal and placental growth at midgestation. To test, on day 12 postbreeding, osmotic pumps were surgically installed in 30 ewes and delivered intrauterine CXCR4 inhibitor or saline for 7 or 14 days. On day 90, fetal/maternal tissues were collected, measured, weighed, and maternal (caruncle) and fetal (cotyledon) placenta components separated and analyzed. The objectives were to determine if (i) suppressing CXCL12/CXCR4 during implantation results in reduced fetal and placental growth and development and (ii) if varying the amount of time CXCL12/CXCR4 is suppressed impacts fetal/placental development. Fetal weights were similar; however greater placental weight and placentome numbers occurred when CXCL12/CXCR4 was suppressed for 14 days. In caruncles, greater abundance of fibroblast growth factor 2, vascular endothelial growth factor A, vascular endothelial growth factor A receptor 1 (FLT-1), and placental growth factor were observed after suppressing CXCL12/CXCR4. Similar results occurred in cotyledons except less vascular endothelial growth factor in 7 day group and less fibroblast growth factor in 14 day group. Our data underscore the importance of CXCL12/CXCR4 signaling during placentation and provide strong evidence that altering CXCL12-mediated signaling induces enduring placental effects manifesting later in gestation.
Collapse
Affiliation(s)
- Ryan L Ashley
- Department of Animal and Range Sciences, New Mexico State University, Las Cruces, NM, USA
| | - Elisa M Trigo
- Department of Animal and Range Sciences, New Mexico State University, Las Cruces, NM, USA
| | - Jacqueline M Ervin
- Department of Animal and Range Sciences, New Mexico State University, Las Cruces, NM, USA
| |
Collapse
|
169
|
Sokratous N, Bednorz M, Syngelaki A, Wright A, Nicolaides KH, Kametas NA. Prediction using serum glycosylated fibronectin and angiogenic factors of superimposed pre-eclampsia in women with chronic hypertension. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2023; 62:836-842. [PMID: 37675881 DOI: 10.1002/uog.27475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/08/2023]
Abstract
OBJECTIVE To compare the predictive performance for delivery with pre-eclampsia (PE) within 2 weeks of assessment in women with chronic hypertension at 24-41 weeks' gestation between serum glycosylated fibronectin (GlyFn) concentration, serum placental growth factor (PlGF) concentration and soluble fms-like tyrosine kinase-1 (sFlt-1) to PlGF concentration ratio. METHODS This was a prospective study of 104 women with a singleton pregnancy and chronic hypertension presenting at 24-41 weeks' gestation. Twenty-six (25.0%) cases developed superimposed PE within 2 weeks of sampling. We compared the predictive performance for superimposed PE between GlyFn, PlGF and the sFlt-1/PlGF ratio at a fixed screen-positive rate of approximately 10%. RESULTS The median gestational age at sampling was 34.1 (interquartile range, 31.5-35.6) weeks and 84.6% (88/104) of cases were sampled at < 36 weeks. The predictive performance for superimposed PE of the three methods of screening was similar, with detection rates of about 23-27%, at a screen-positive rate of 11% and a false-positive rate of about 5%. CONCLUSIONS Measurement of GlyFn is a simple point-of-care test that can be carried out without need for a laboratory and provide results within 10 min of testing. In this respect, it could potentially replace the angiogenic markers that are used currently in the prediction of imminent PE in high-risk women. However, neither GlyFn nor angiogenic factors are likely to improve the management of women with chronic hypertension because their predictive performance for superimposed PE is poor. © 2023 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- N Sokratous
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - M Bednorz
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - A Syngelaki
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - A Wright
- Institute of Health Research, University of Exeter, Exeter, UK
| | - K H Nicolaides
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - N A Kametas
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| |
Collapse
|
170
|
Hackelöer M, Schmidt L, Verlohren S. New advances in prediction and surveillance of preeclampsia: role of machine learning approaches and remote monitoring. Arch Gynecol Obstet 2023; 308:1663-1677. [PMID: 36566477 PMCID: PMC9790089 DOI: 10.1007/s00404-022-06864-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/18/2022] [Indexed: 12/26/2022]
Abstract
Preeclampsia, a multisystem disorder in pregnancy, is still one of the main causes of maternal morbidity and mortality. Due to a lack of a causative therapy, an accurate prediction of women at risk for the disease and its associated adverse outcomes is of utmost importance to tailor care. In the past two decades, there have been successful improvements in screening as well as in the prediction of the disease in high-risk women. This is due to, among other things, the introduction of biomarkers such as the sFlt-1/PlGF ratio. Recently, the traditional definition of preeclampsia has been expanded based on new insights into the pathophysiology and conclusive evidence on the ability of angiogenic biomarkers to improve detection of preeclampsia-associated maternal and fetal adverse events.However, with the widespread availability of digital solutions, such as decision support algorithms and remote monitoring devices, a chance for a further improvement of care arises. Two lines of research and application are promising: First, on the patient side, home monitoring has the potential to transform the traditional care pathway. The importance of the ability to input and access data remotely is a key learning from the COVID-19 pandemic. Second, on the physician side, machine-learning-based decision support algorithms have been shown to improve precision in clinical decision-making. The integration of signals from patient-side remote monitoring devices into predictive algorithms that power physician-side decision support tools offers a chance to further improve care.The purpose of this review is to summarize the recent advances in prediction, diagnosis and monitoring of preeclampsia and its associated adverse outcomes. We will review the potential impact of the ability to access to clinical data via remote monitoring. In the combination of advanced, machine learning-based risk calculation and remote monitoring lies an unused potential that allows for a truly patient-centered care.
Collapse
Affiliation(s)
- Max Hackelöer
- Department of Obstetrics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Leon Schmidt
- Department of Obstetrics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Stefan Verlohren
- Department of Obstetrics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
171
|
Kumar M, Balyan K, Debnath E, Himtsoe B, Sapna S, Kumar A. Antenatal risk stratification for preeclampsia with sFlt-1/PlGF ratio: Which is the best time to test? Pregnancy Hypertens 2023; 34:39-46. [PMID: 37801800 DOI: 10.1016/j.preghy.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 07/15/2023] [Accepted: 09/21/2023] [Indexed: 10/08/2023]
Abstract
OBJECTIVE To find out the predictive value of sFlt-1/PlGF ratio for antenatal risk stratification (ARS) of women at high risk of preeclampsia (PE). METHODS Antenatal women at high risk of PE underwent sFlt-1/PlGF ratio at 20-22, 28-30 and 34-36 weeks and were followed till delivery. Those who developed PE were cases those who had normal outcome were controls, the cases and controls were compared. RESULTS Hypertension in pregnancy was seen in 116/287 (40.4 %), 46/287(16.0 %) had PE and 21(7.3 %) had early onset PE. Mean arterial pressure at 20-22 weeks was the high in those who developed early onset PE (109.08 ± 9.74 mmHg). The sFlt-1/PlGF ratio of 38 or more at 20-22 weeks resulted in either PE or adverse fetal outcome in all cases. Whereas, the ratio of less than 38 ruled out PE in all cases up to 29 + 6 weeks. At 28-30 weeks, the ratio less than 38 predicted no PE up to 34 weeks and no complication up to 29+6 weeks. The sensitivity for the detection at later gestation further decreased as the gestation advanced however the specificity was above 98 % at all gestations. The positive predictive value of the test increased with the advancing gestation, the negative predictive value was 93 % or higher at all gestations. CONCLUSION The usefulness of sFlt-1/PlGF ratio ≥38 for risk stratification was validated in the study, the testing at 28-30 weeks appeared to be the best time to test for PE prediction in high risk women.
Collapse
Affiliation(s)
- Manisha Kumar
- Department of Obstetrics and Gynecology, LHMC, New Delhi, India.
| | - Kirti Balyan
- Department of Obstetrics and Gynecology, LHMC, New Delhi, India
| | - Ekta Debnath
- Department of Biochemistry, LHMC, New Delhi, India
| | - Ben Himtsoe
- Department of Obstetrics and Gynecology, LHMC, New Delhi, India
| | - Surbhi Sapna
- Department of Obstetrics and Gynecology, LHMC, New Delhi, India
| | - Arunav Kumar
- Kasturba Medical College, Manipal, Karnataka, India
| |
Collapse
|
172
|
Binder J, Palmrich P, Kalafat E, Haberl C, Schirwani N, Pateisky P, Khalil A. Longitudinal assessment of angiogenic markers in prediction of adverse outcome in women with confirmed pre-eclampsia. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2023; 62:843-851. [PMID: 37265117 DOI: 10.1002/uog.26276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 06/03/2023]
Abstract
OBJECTIVES Angiogenic marker assessment, such as the ratio of soluble fms-like tyrosine kinase-1 (sFlt-1) to placental growth factor (PlGF), is known to be a useful tool in the prediction of pre-eclampsia (PE). However, evidence from surveillance strategies in pregnancies with a PE diagnosis is lacking. Therefore, we aimed to assess the predictive performance of longitudinal maternal serum angiogenic marker assessment for both maternal and perinatal adverse outcomes when compared to standard laboratory parameters in pregnancies with confirmed PE. METHODS This was a retrospective analysis of prospectively collected data from January 2013 to December 2020 at the Medical University of Vienna. The inclusion criteria were singleton pregnancy with confirmed PE and post-diagnosis maternal serum angiogenic marker assessment at a minimum of two timepoints. The primary outcome was the predictive performance of longitudinal sFlt-1 and PlGF assessment for adverse maternal and perinatal outcomes compared to conventional laboratory monitoring at the same time in pregnancies with confirmed PE. Composite adverse maternal outcome included intensive care unit admission, pulmonary edema, eclampsia and/or death. Composite adverse perinatal outcome included stillbirth, neonatal death, placental abruption, neonatal intensive care unit admission, intraventricular hemorrhage, necrotizing enterocolitis, respiratory distress syndrome and/or mechanical ventilator support. RESULTS In total, 885 post-diagnosis sFlt-1/PlGF ratio measurements were obtained from 323 pregnant women with confirmed PE. For composite adverse maternal outcome, the highest standalone predictive accuracy was obtained using maternal serum sFlt-1/PlGF ratio (area under the receiver-operating-characteristics curve (AUC), 0.72 (95% CI, 0.62-0.81)), creatinine (AUC, 0.71 (95% CI, 0.62-0.81)) and lactate dehydrogenase (LDH) levels (AUC, 0.73 (95% CI, 0.65-0.81)). Maternal platelet levels (AUC, 0.65 (95% CI, 0.55-0.74)), serum alanine aminotransferase (ALT) (AUC, 0.59 (95% CI, 0.49-0.69)) and aspartate aminotransferase (AST) (AUC, 0.61 (95% CI, 0.51-0.71) levels had poor standalone predictive accuracy. The best prediction model consisted of a combination of maternal serum LDH, creatinine levels and sFlt-1/PlGF ratio, which had an AUC of 0.77 (95% CI, 0.68-0.85), significantly higher than sFlt-1/PlGF ratio alone (P = 0.037). For composite adverse perinatal outcome, the highest standalone predictive accuracy was obtained using maternal serum sFlt-1/PlGF ratio (AUC, 0.82 (95% CI, 0.75-0.89)) and creatinine (AUC, 0.74 (95% CI, 0.67-0.80)) levels, sFlt-1/PlGF ratio being superior to creatinine alone (P < 0.001). Maternal serum LDH levels (AUC, 0.65 (95% CI, 0.53-0.74)), platelet count (AUC, 0.57 (95% CI, 0.44-0.67)), ALT (AUC, 0.58 (95% CI, 0.48-0.67)) and AST (AUC, 0.58 (95% CI, 0.48-0.67)) levels had poor standalone predictive accuracy. No combination of biomarkers was superior to maternal serum sFlt-1/PlGF ratio alone for prediction of composite adverse perinatal outcome (P > 0.05 for all). CONCLUSIONS In pregnancies with confirmed PE, longitudinal maternal serum angiogenic marker assessment is a good predictor of adverse maternal and perinatal outcomes and superior to some conventional laboratory parameters. Further studies should focus on optimal surveillance following diagnosis of PE. © 2023 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- J Binder
- Department of Obstetrics and Fetomaternal Medicine, Medical University of Vienna, Vienna, Austria
| | - P Palmrich
- Department of Obstetrics and Fetomaternal Medicine, Medical University of Vienna, Vienna, Austria
| | - E Kalafat
- Department of Obstetrics and Gynecology, School of Medicine, Koc University, Istanbul, Turkey
| | - C Haberl
- Department of Obstetrics and Fetomaternal Medicine, Medical University of Vienna, Vienna, Austria
| | - N Schirwani
- Department of Obstetrics and Fetomaternal Medicine, Medical University of Vienna, Vienna, Austria
| | - P Pateisky
- Department of Obstetrics and Fetomaternal Medicine, Medical University of Vienna, Vienna, Austria
| | - A Khalil
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust, University of London, London, UK
- Vascular Biology Research Centre, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| |
Collapse
|
173
|
Lau KGY, Kountouris E, Salazar-Rios L, Nicolaides KH, Kametas NA. Prediction of adverse outcome by ophthalmic artery Doppler and angiogenic markers in pregnancies with new onset hypertension. Pregnancy Hypertens 2023; 34:110-115. [PMID: 37925874 DOI: 10.1016/j.preghy.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 07/31/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023]
Abstract
OBJECTIVES To compare the ophthalmic artery Doppler peak systolic velocity ratio (OA PSV-ratio) and soluble fms-like tyrosine kinase-1/placental growth factor ratio (sFlt-1/PlGF ratio) in predicting adverse maternal and perinatal outcomes in women presenting with new onset hypertension. STUDY DESIGN Prospective cohort study in a specialist hypertension clinic, within a tertiary referral centre. MAIN OUTCOME MEASURES Comparison between the OA PSV-ratio and sFlt-1/PlGF ratio in predicting delivery within one week from presentation and adverse maternal and perinatal outcomes e.g. severe hypertension, neonatal unit admission, small for gestational age. RESULTS Women who delivered within one week, compared to those who did not, had a higher OA PSV-ratio (0.82 vs 0.71, p < 0.01) and sFlt-1/PlGF ratio (93.3 vs 40.5, p = 0.08). Independent predictors of the OA PSV-ratio included mean arterial pressure and maternal weight and predictors of the sFlt-1/PlGF ratio included diastolic blood pressure and use of antihypertensive medications. Prediction of adverse outcomes with both ratios were similar and only modest e.g. AUROC for predicting delivery within one week for OA PSV-ratio was 0.57 (95% CI 0.47-0.67) and for sFlt-1/PlGF ratio was 0.61 (95% CI 0.52-0.70) (p = 0.53). CONCLUSIONS In women presenting with new onset hypertension, the OA PSV-ratio and sFlt-1/PlGF ratio have similar and modest performance in predicting adverse outcomes.
Collapse
Affiliation(s)
- Katherine G Y Lau
- Antenatal Hypertension Clinic, King's College Hospital, London, UK; Harris Birthright, Research Centre for Fetal Medicine, King's College Hospital, London, UK
| | | | | | - Kypros H Nicolaides
- Harris Birthright, Research Centre for Fetal Medicine, King's College Hospital, London, UK
| | - Nikos A Kametas
- Antenatal Hypertension Clinic, King's College Hospital, London, UK; Harris Birthright, Research Centre for Fetal Medicine, King's College Hospital, London, UK.
| |
Collapse
|
174
|
Álvarez SC, Zurita AR, Del Carmen De Ganzo Suárez T, De Luis Escudero JF, Medina NS, Pérez CC, de Basoa CMF, Montesino JLT, Masip MTC, Bello MÁG. Is a sFlt-1/PlGF cutoff of 38 suitable to predict adverse outcomes in pregnancies with abnormal uterine artery Doppler velocimetry in the second trimester? Pregnancy Hypertens 2023; 34:13-18. [PMID: 37778280 DOI: 10.1016/j.preghy.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 07/03/2023] [Accepted: 09/21/2023] [Indexed: 10/03/2023]
Abstract
OBJECTIVE To determine the optimal cutoff value for the soluble fms-like tyrosine kinase-1 (sFlt-1)/placental growth factor (PlGF) ratio to predict maternal and fetal adverse events in pregnancies with uterine artery Doppler scans results above the 95th percentile in the late second trimester. STUDY DESIGN Retrospective, observational cohort study on 116 asyntomatic patients with abnormal uterine artery Doppler scans at gestational week 25. The sFlt-1/PlGF ratio was determined within the weeks 25 to 29 of gestation and ROC curve analysis performed. The diagnostic validity of different cutoff values to predict severe maternal and fetal complications, i.e. preeclampsia, fetal growth restriction, placental abruption, and fetal death, was analyzed. MAIN OUTCOME MEASURES An ideal cutoff for sFlt-1/PlGF ratios in pregnancies with abnormal uterine artery Doppler in the second trimester. RESULTS Applying a cutoff point of 38, the area under the ROC curve was 0.89, generally considered low risk in fetal and maternal complication prediction. The sensitivity was 32.1%, the specificity 98.4%, the positive predictive value (PPV) 94.4%, and the negative predictive value (NPV) 63.3%. A cutoff value of 10, leading to the highest Youden index, performed best at detecting overall complications, increasing sensitivity to 69.8% and the NPV to 76.8%. at the cost of a reduced specificity and PPV. CONCLUSIONS In pregnancies with abnormal uterine artery Doppler in the second trimester, an sFlt-1/PlGF cutoff value greater than equal to 38 improves its predictive power for adverse events.
Collapse
Affiliation(s)
- Sara Caamiña Álvarez
- Department of Obstetrics and Gynecology, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain.
| | - Alicia Rodríguez Zurita
- Department of Obstetrics and Gynecology, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | | | - José Fernando De Luis Escudero
- Department of Obstetrics and Gynecology, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Nieves Sierra Medina
- Department of Obstetrics and Gynecology, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Carolina Chulilla Pérez
- Department of Obstetrics and Gynecology, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Cecilia Martín Fernández de Basoa
- Prenatal Screening and Preeclampsia Unit, Clinical Analysis Service, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - José Luis Trabado Montesino
- Department of Obstetrics and Gynecology, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - María Teresa Concepción Masip
- Prenatal Screening and Preeclampsia Unit, Clinical Analysis Service, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Miguel Ángel García Bello
- Department of Clinical Psychology-Psychobiology and Methodology, University of La Laguna, Santa Cruz de Tenerife, Spain
| |
Collapse
|
175
|
Aisagbonhi O, Bui T, Nasamran CA, St Louis H, Pizzo D, Meads M, Mulholland M, Magallanes C, Lamale-Smith L, Laurent LC, Morey R, Jacobs MB, Fisch KM, Horii M. High placental expression of FLT1, LEP, PHYHIP and IL3RA - In persons of African ancestry with severe preeclampsia. Placenta 2023; 144:13-22. [PMID: 37949031 PMCID: PMC10843761 DOI: 10.1016/j.placenta.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023]
Abstract
INTRODUCTION Mortality from preeclampsia (PE) and PE-associated morbidities are 3-to 5-fold higher in persons of African ancestry than in those of Asian and European ancestries. METHODS To elucidate placental contribution to worse PE outcomes in African ancestry pregnancies, we performed bulk RNA sequencing on 50 placentas from persons with severe PE (sPE) of African (n = 9), Asian (n = 18) and European (n = 23) ancestries and 73 normotensive controls of African (n = 10), Asian (n = 15) and European (n = 48) ancestries. RESULTS Previously described canonical preeclampsia genes, involved in metabolism and hypoxia/angiogenesis including: LEP, HK2, FSTL3, FLT1, ENG, TMEM45A, ARHGEF4 and HTRA1 were upregulated sPE versus normotensive placentas across ancestries. LTF, NPR3 and PHYHIP were higher in African vs. Asian ancestry sPE placentas. Allograft rejection/adaptive immune response genes were upregulated in placentas from African but not in Asian or European ancestry sPE patients; IL3RA was of particular interest because the patient with the highest placental IL3RA expression, a person of African ancestry with sPE, developed postpartum cardiomyopathy, and was the only patient out of 123, that developed this condition. Interestingly, the sPE patients with the highest IL3RA expression among persons of Asian and European ancestries developed unexplained tachycardia peripartum, necessitating echocardiography in the European ancestry patient. The association between elevated placental IL3RA levels and unexplained tachycardia or peripartum cardiomyopathy was found to be significant in the 50 sPE patients (p = .0005). DISCUSSION High placental upregulation of both canonical preeclampsia and allograft rejection/adaptive immune response genes may contribute to worse PE outcomes in African ancestry sPE patients.
Collapse
Affiliation(s)
- Omonigho Aisagbonhi
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Center for Perinatal Discovery, University of California, San Diego, La Jolla, CA, USA.
| | - Tony Bui
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Center for Perinatal Discovery, University of California, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Chanond A Nasamran
- Center for Computational Biology and Bioinformatics, University of California, San Diego, La Jolla, CA, USA
| | - Hailee St Louis
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Donald Pizzo
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Morgan Meads
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Center for Perinatal Discovery, University of California, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Megan Mulholland
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Center for Perinatal Discovery, University of California, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Celestine Magallanes
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Leah Lamale-Smith
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Louise C Laurent
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Robert Morey
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Marni B Jacobs
- Center for Perinatal Discovery, University of California, San Diego, La Jolla, CA, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Kathleen M Fisch
- Center for Perinatal Discovery, University of California, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA; Center for Computational Biology and Bioinformatics, University of California, San Diego, La Jolla, CA, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Mariko Horii
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Center for Perinatal Discovery, University of California, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
176
|
Pan HY, Wang TD. The association of a panel of circulating markers with hypertensive disorders of pregnancy: the jury is still out. Hypertens Res 2023; 46:2759-2761. [PMID: 37821567 DOI: 10.1038/s41440-023-01445-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 10/13/2023]
Abstract
Comparison between using either sFlt-1/PlGF ratio or proposed panel of biomarkers. The latter is proposed by using statistical and machine learning methods. The levels of both sFlt-1 and PlGF are measured in pg/mL. sFlt-1 soluble fms-like tyrosine kinase-1, PlGF placental growth factor, PPV positive predictive value, NPV negative predictive value.
Collapse
Affiliation(s)
- Heng-Yu Pan
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Hsinchu Branch, Hsinchu City, Taiwan
| | - Tzung-Dau Wang
- Divisions of Cardiology and Hospital Medicine, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan.
| |
Collapse
|
177
|
He W, Zhang Y, Wu K, Wang Y, Zhao X, Lv L, Ren C, Lu J, Yang J, Yin A, Liu G. Epigenetic phenotype of plasma cell-free DNA in the prediction of early-onset preeclampsia. J OBSTET GYNAECOL 2023; 43:2282100. [PMID: 38038254 DOI: 10.1080/01443615.2023.2282100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 11/06/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUND In the current study, we sought to characterise the methylation haplotypes and nucleosome positioning patterns of placental DNA and plasma cell-free DNA of pregnant women with early-onset preeclampsia using whole genome bisulphite sequencing (WGBS) and methylation capture bisulphite sequencing (MCBS) and further develop and examine the diagnostic performance of a generalised linear model (GLM) by incorporating the epigenetic features for early-onset preeclampsia. METHODS This case-control study recruited pregnant women aged at least 18 years who delivered their babies at our Hospital. In addition, non-pregnant women with no previous history of diseases were included. Placental samples of the villous parenchyma were taken at the time of delivery and venous blood was drawn from pregnant women during non-invasive prenatal testing at 12-15 weeks of pregnancy and nonpregnant women during the physical check-up. WGBS and MCBS were carried out of extracted genomic DNA. Then, we established the GLM by incorporating preeclampsia-specific methylation haplotypes and nucleosome positioning patterns and examined the diagnostic performance of the model by receiver operating characteristic (ROC) curve analysis. RESULTS The study included 135 pregnant women and 50 non-pregnant women. Our high-depth MCBS revealed notably different DNA methylation and nucleosome positioning patterns between women with and without preeclampsia. Preeclampsia-specific hypermethylated sites were found predominantly in the promoter regions and particularly enriched in CTCF on the X chromosome. Totally, 2379 preeclampsia-specific methylation haplotypes were found across the entire genome. ROC analysis showed that the area under the ROC curve (AUC) was 0.938 (95%CI 0.877, 1.000). At a GLM cut-off of 0.341, the AUC was the maximum, with a sensitivity of 95.6% and a specificity of 89.7%. CONCLUSION Pregnant women with early-onset preeclampsia exhibit DNA methylation and nucleosome positioning patterns in placental and plasma DNA.
Collapse
Affiliation(s)
- Wei He
- The First Affiliated Hospital of Jinan University, Guangzhou, China
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Yi Zhang
- Euler Technology, Beijing, China
- Peking-Tsinghua Center of Life Sciences, Beijing, China
- School of Life Sciences, Peking University, Beijing, China
| | - Kai Wu
- Euler Technology, Beijing, China
| | - Yunan Wang
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Xin Zhao
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Lijuan Lv
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Congmian Ren
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Jiaqi Lu
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Jiexia Yang
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Aihua Yin
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Guocheng Liu
- Department of Obstetrics, Guangdong Women and Children Hospital, Guangzhou, China
| |
Collapse
|
178
|
Palomo M, Moreno-Castaño AB, Salas MQ, Escribano-Serrat S, Rovira M, Guillen-Olmos E, Fernandez S, Ventosa-Capell H, Youssef L, Crispi F, Nomdedeu M, Martinez-Sanchez J, De Moner B, Diaz-Ricart M. Endothelial activation and damage as a common pathological substrate in different pathologies and cell therapy complications. Front Med (Lausanne) 2023; 10:1285898. [PMID: 38034541 PMCID: PMC10682735 DOI: 10.3389/fmed.2023.1285898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
The endothelium is a biologically active interface with multiple functions, some of them common throughout the vascular tree, and others that depend on its anatomical location. Endothelial cells are continually exposed to cellular and humoral factors, and to all those elements (biological, chemical, or hemodynamic) that circulate in blood at a certain time. It can adapt to different stimuli but this capability may be lost if the stimuli are strong enough and/or persistent in time. If the endothelium loses its adaptability it may become dysfunctional, becoming a potential real danger to the host. Endothelial dysfunction is present in multiple clinical conditions, such as chronic kidney disease, obesity, major depression, pregnancy-related complications, septic syndromes, COVID-19, and thrombotic microangiopathies, among other pathologies, but also in association with cell therapies, such as hematopoietic stem cell transplantation and treatment with chimeric antigen receptor T cells. In these diverse conditions, evidence suggests that the presence and severity of endothelial dysfunction correlate with the severity of the associated disease. More importantly, endothelial dysfunction has a strong diagnostic and prognostic value for the development of critical complications that, although may differ according to the underlying disease, have a vascular background in common. Our multidisciplinary team of women has devoted many years to exploring the role of the endothelium in association with the mentioned diseases and conditions. Our research group has characterized some of the mechanisms and also proposed biomarkers of endothelial damage. A better knowledge would provide therapeutic strategies either to prevent or to treat endothelial dysfunction.
Collapse
Affiliation(s)
- Marta Palomo
- Hemostasis and Erythropathology Laboratory, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Institut de Recerca August Pi Sunyer, University of Barcelona, Barcelona, Spain
- Hematology External Quality Assessment Laboratory, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Ana Belén Moreno-Castaño
- Hemostasis and Erythropathology Laboratory, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Institut de Recerca August Pi Sunyer, University of Barcelona, Barcelona, Spain
| | - María Queralt Salas
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Cancer and Blood Diseases, Hospital Clínic de Barcelona, Institut de Recerca August Pi Sunyer, Barcelona, Spain
| | - Silvia Escribano-Serrat
- Hemostasis and Erythropathology Laboratory, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Institut de Recerca August Pi Sunyer, University of Barcelona, Barcelona, Spain
| | - Montserrat Rovira
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Cancer and Blood Diseases, Hospital Clínic de Barcelona, Institut de Recerca August Pi Sunyer, Barcelona, Spain
| | - Elena Guillen-Olmos
- Department of Nephrology and Kidney Transplantation, Hospital Clínic de Barcelona, Centro de Referencia en Enfermedad Glomerular Compleja del Sistema Nacional de Salud (CSUR), University of Barcelona, Barcelona, Spain
| | - Sara Fernandez
- Medical Intensive Care Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - Lina Youssef
- BCNatal – Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic de Barcelona and Hospital Sant Joan de Déu, Institut de Recerca August Pi Sunyer, University of Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Fatima Crispi
- BCNatal – Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic de Barcelona and Hospital Sant Joan de Déu, Institut de Recerca August Pi Sunyer, University of Barcelona, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| | - Meritxell Nomdedeu
- Hemostasis and Hemotherapy Department, Institute of Cancer and Blood Diseases, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Julia Martinez-Sanchez
- Hemostasis and Erythropathology Laboratory, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Institut de Recerca August Pi Sunyer, University of Barcelona, Barcelona, Spain
| | - Blanca De Moner
- Hemostasis and Erythropathology Laboratory, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Institut de Recerca August Pi Sunyer, University of Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Maribel Diaz-Ricart
- Hemostasis and Erythropathology Laboratory, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Institut de Recerca August Pi Sunyer, University of Barcelona, Barcelona, Spain
| |
Collapse
|
179
|
Bruno V, Barth D, Jauhal A. Complement-Mediated Thrombotic Microangiopathy in Pregnancy: An Educational Case Report. Can J Kidney Health Dis 2023; 10:20543581231209009. [PMID: 37942411 PMCID: PMC10629310 DOI: 10.1177/20543581231209009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/06/2023] [Indexed: 11/10/2023] Open
Abstract
Rationale Thrombotic microangiopathy (TMA) is a spectrum of rare diseases characterized by thrombocytopenia, microangiopathic hemolytic anemia, and organ damage. Differentiating pre-eclampsia, HELLP (Hemolysis, Elevated Liver enzymes, Low Platelets) syndrome and atypical hemolytic uremic syndrome (aHUS) during pregnancy may be diagnostically challenging yet important as the treatment pathways differ. Most cases of aHUS are associated with dysregulation of the complement alternative pathway, for which current guidelines recommend prompt treatment with complement C5 inhibitor to prevent chronic sequelae. Here, we report a case of pregnancy-associated aHUS (p-aHUS) to highlight the challenging aspects of the diagnostic process and the importance of prompt treatment with complement inhibition to reduce the risk of poor outcomes. Presenting concerns A 28-year-old woman was admitted to a local hospital for induction of vaginal delivery of twins at 34 weeks and 3 days of gestational age, due to intrauterine growth restriction (IUGR). She was previously healthy, and this current pregnancy was uncomplicated, except for the IUGR. Approximately, 10 hours after her induced delivery, she developed vomiting, epigastric pain, and hypertension. Diagnosis She was initially suspected of having fulminant liver failure in the context of acute fatty liver of pregnancy versus pre-eclampsia/HELLP syndrome, due to evidence of elevated liver enzymes, acute kidney injury (AKI), thrombocytopenia, and hemoglobin levels trending down, for which the patient was initially treated conservatively. On day 2 post-delivery, she was transferred to our hospital for possible liver biopsy and management of liver failure. Upon transfer, dialysis was started due to anuric AKI; at the same time, her liver function spontaneously improved, while platelet count remained very low and hemoglobin levels continued to trend down. A full TMA work-up revealed low C3 levels; secondary causes of TMA were ruled out. The patient received a final diagnosis of p-aHUS. Complement genetic tests were also performed and did not identify any pathogenic variants. Interventions Given the final diagnosis of p-aHUS, the patient was started on a C5 inhibitor (day 8 post-delivery). Her platelet count quickly normalized 2 days after the first dose, while the hemoglobin levels remained low for a longer period, likely due to retained products of conception. Outcomes The patient was able to completely discontinue dialysis after approximately 3 months, however, her kidney function did not recover completely, despite all the other TMA markers normalizing (platelets count in range, negative hemolysis markers, and normal hemoglobin levels). Her estimated glomerular filtration rate (eGFR) was 23 mL/min/1.73 m2 at the 6-month follow-up. Teaching points The diagnosis of p-aHUS can be challenging due to frequent overlapping symptoms and signs with other forms of pregnancy-associated TMA, leading to a delay of the treatment, which can affect the patient's outcome. Failure of TMA to improve in the postpartum period or occurring at this time, with negative ADAMTS13 and antiphospholipid antibody syndrome (APLAS) serologies should favor the diagnosis of p-aHUS. Early treatment with C5 inhibition should be considered in women with a diagnosis of p-aHUS. Patients need multidisciplinary and likely tertiary/quaternary care at centers where clinical experience, access to diagnostics and treatment initiation can begin without delay.
Collapse
Affiliation(s)
- Valentina Bruno
- Division of Nephrology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, ON, Canada
- Cell Biology Program, SickKids Research Institute, The Hospital for Sick Children, University of Toronto, ON, Canada
| | - David Barth
- Division of Medical Oncology and Hematology, University Health Network, Temerty Faculty of Medicine, University of Toronto, ON, Canada
- Departments of Medicine and Laboratory Medicine & Pathobiology, University of Toronto, ON, Canada
| | - Arenn Jauhal
- Division of Nephrology, Department of Medicine, Glomerulonephritis Clinic, Toronto General Hospital, University Health Network, Temerty Faculty of Medicine, University of Toronto, ON, Canada
| |
Collapse
|
180
|
Zhang Q, Lee CL, Yang T, Li J, Zeng Q, Liu X, Liu Z, Ruan D, Li Z, Kan AS, Cheung KW, Mak AS, Ng VW, Zhao H, Fan X, Duan YG, Zhong L, Chen M, Du M, Li RH, Liu P, Ng EH, Yeung WS, Gao Y, Yao Y, Chiu PC. Adrenomedullin has a pivotal role in trophoblast differentiation: A promising nanotechnology-based therapeutic target for early-onset preeclampsia. SCIENCE ADVANCES 2023; 9:eadi4777. [PMID: 37922358 PMCID: PMC10624351 DOI: 10.1126/sciadv.adi4777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 10/03/2023] [Indexed: 11/05/2023]
Abstract
Early-onset preeclampsia (EOPE) is a severe pregnancy complication associated with defective trophoblast differentiation and functions at implantation, but manifestation of its phenotypes is in late pregnancy. There is no reliable method for early prediction and treatment of EOPE. Adrenomedullin (ADM) is an abundant placental peptide in early pregnancy. Integrated single-cell sequencing and spatial transcriptomics confirm a high ADM expression in the human villous cytotrophoblast and syncytiotrophoblast. The levels of ADM in chorionic villi and serum were lower in first-trimester pregnant women who later developed EOPE than those with normotensive pregnancy. ADM stimulates differentiation of trophoblast stem cells and trophoblast organoids in vitro. In pregnant mice, placenta-specific ADM suppression led to EOPE-like phenotypes. The EOPE-like phenotypes in a mouse PE model were reduced by a placenta-specific nanoparticle-based forced expression of ADM. Our study reveals the roles of trophoblastic ADM in placental development, EOPE pathogenesis, and its potential clinical uses.
Collapse
Affiliation(s)
- Qingqing Zhang
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Cheuk-Lun Lee
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Tingyu Yang
- BGI-Shenzhen, Shenzhen 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianlin Li
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Qunxiong Zeng
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Xiaofeng Liu
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zhongzhen Liu
- BGI-Shenzhen, Shenzhen 518083, China
- Shenzhen Engineering Laboratory for Birth Defects Screening, Shenzhen, China
| | - Degong Ruan
- Stem Cell and Regenerative Medicine Consortium, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhuoxuan Li
- Stem Cell and Regenerative Medicine Consortium, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Anita S. Y. Kan
- Department of Obstetrics and Gynaecology, Queen Mary Hospital, Hong Kong, China
| | - Ka-Wang Cheung
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Annisa S. L. Mak
- Department of Obstetrics and Gynaecology, Queen Elizabeth Hospital, Hong Kong, China
| | - Vivian W. Y. Ng
- Department of Obstetrics and Gynaecology, Queen Mary Hospital, Hong Kong, China
| | - Huashan Zhao
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiujun Fan
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yong-Gang Duan
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Liuying Zhong
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Min Chen
- Department of Prenatal Diagnosis and Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Meirong Du
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Raymond H. W. Li
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Pengtao Liu
- Stem Cell and Regenerative Medicine Consortium, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ernest H. Y. Ng
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - William S. B. Yeung
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Ya Gao
- BGI-Shenzhen, Shenzhen 518083, China
- Shenzhen Engineering Laboratory for Birth Defects Screening, Shenzhen, China
| | - Yuanqing Yao
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Philip C. N. Chiu
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
181
|
Omosule CL. Prognostication of Preeclampsia. Clin Chem 2023; 69:1324-1325. [PMID: 37932108 DOI: 10.1093/clinchem/hvad123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 07/25/2023] [Indexed: 11/08/2023]
Affiliation(s)
- Catherine L Omosule
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
182
|
Parisi F, Fenizia C, Introini A, Zavatta A, Scaccabarozzi C, Biasin M, Savasi V. The pathophysiological role of estrogens in the initial stages of pregnancy: molecular mechanisms and clinical implications for pregnancy outcome from the periconceptional period to end of the first trimester. Hum Reprod Update 2023; 29:699-720. [PMID: 37353909 PMCID: PMC10628507 DOI: 10.1093/humupd/dmad016] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 05/12/2023] [Indexed: 06/25/2023] Open
Abstract
BACKGROUND Estrogens regulate disparate female physiological processes, thus ensuring reproduction. Altered estrogen levels and signaling have been associated with increased risks of pregnancy failure and complications, including hypertensive disorders and low birthweight babies. However, the role of estrogens in the periconceptional period and early pregnancy is still understudied. OBJECTIVE AND RATIONALE This review aims to summarize the current evidence on the role of maternal estrogens during the periconceptional period and the first trimester of pregnancies conceived naturally and following ART. Detailed molecular mechanisms and related clinical impacts are extensively described. SEARCH METHODS Data for this narrative review were independently identified by seven researchers on Pubmed and Embase databases. The following keywords were selected: 'estrogens' OR 'estrogen level(s)' OR 'serum estradiol' OR 'estradiol/estrogen concentration', AND 'early pregnancy' OR 'first trimester of pregnancy' OR 'preconceptional period' OR 'ART' OR 'In Vitro Fertilization (IVF)' OR 'Embryo Transfer' OR 'Frozen Embryo Transfer' OR 'oocyte donation' OR 'egg donation' OR 'miscarriage' OR 'pregnancy outcome' OR 'endometrium'. OUTCOMES During the periconceptional period (defined here as the critical time window starting 1 month before conception), estrogens play a crucial role in endometrial receptivity, through the activation of paracrine/autocrine signaling. A derailed estrogenic milieu within this period seems to be detrimental both in natural and ART-conceived pregnancies. Low estrogen levels are associated with non-conception cycles in natural pregnancies. On the other hand, excessive supraphysiologic estrogen concentrations at time of the LH peak correlate with lower live birth rates and higher risks of pregnancy complications. In early pregnancy, estrogen plays a massive role in placentation mainly by modulating angiogenic factor expression-and in the development of an immune-tolerant uterine micro-environment by remodeling the function of uterine natural killer and T-helper cells. Lower estrogen levels are thought to trigger abnormal placentation in naturally conceived pregnancies, whereas an estrogen excess seems to worsen pregnancy development and outcomes. WIDER IMPLICATIONS Most current evidence available endorses a relation between periconceptional and first trimester estrogen levels and pregnancy outcomes, further depicting an optimal concentration range to optimize pregnancy success. However, how estrogens co-operate with other factors in order to maintain a fine balance between local tolerance towards the developing fetus and immune responses to pathogens remains elusive. Further studies are highly warranted, also aiming to identify the determinants of estrogen response and biomarkers for personalized estrogen administration regimens in ART.
Collapse
Affiliation(s)
- F Parisi
- Department of Woman, Mother and Neonate, ‘V. Buzzi’ Children Hospital, ASST Fatebenefratelli Sacco, Milan, via L. Castelvetro 32, Milan, Italy
| | - C Fenizia
- Department of Pathophysiology and Transplantation, University of Milan, Milan, via F. Sforza 35, Milan 20122, Italy
- Department of Biomedical and Clinical Sciences, “L.Sacco” Hospital, University of Milan, Milan, via G.B. Grassi 74, Milan 20157, Italy
| | - A Introini
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Nobels väg 5, Stockholm, Sweden
| | - A Zavatta
- Department of Woman, Mother and Neonate, ‘V. Buzzi’ Children Hospital, ASST Fatebenefratelli Sacco, Milan, via L. Castelvetro 32, Milan, Italy
| | - C Scaccabarozzi
- Department of Biomedical and Clinical Sciences, “L.Sacco” Hospital, University of Milan, Milan, via G.B. Grassi 74, Milan 20157, Italy
| | - M Biasin
- Department of Biomedical and Clinical Sciences, “L.Sacco” Hospital, University of Milan, Milan, via G.B. Grassi 74, Milan 20157, Italy
| | - V Savasi
- Department of Biomedical and Clinical Sciences, “L.Sacco” Hospital, University of Milan, Milan, via G.B. Grassi 74, Milan 20157, Italy
| |
Collapse
|
183
|
He L, Wu X, Zhan F, Li X, Wu J. Protective role of metformin in preeclampsia via the regulation of NF-κB/sFlt-1 and Nrf2/HO-1 signaling pathways by activating AMPK. Placenta 2023; 143:91-99. [PMID: 37866322 DOI: 10.1016/j.placenta.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/11/2023] [Accepted: 10/07/2023] [Indexed: 10/24/2023]
Abstract
INTRODUCTION Preeclampsia (PE) is a pregnancy complication that leads to hypertension and proteinuria and causes maternal mortality. Metformin (MET) is an oral hypoglycemic agent that activates AMPK-regulated signaling pathways and inhibits inflammation and oxidative stress responses. This study explored MET's roles and molecular mechanisms in PE. METHODS The protein or mRNA expression of signaling pathways and inflammation-related genes were detected by Western blotting and RT-qPCR and cell viability was analyzed with MTT. In addition, flow cytometry was used to assess apoptosis, and mitochondrial membrane potential was detected using JC-1 staining with flow cytometry. Moreover, LDH Cytotoxicity Assay Kit detected the release of LDH, and ROS, MDA, or SOD kits detected oxidative stress-related factors. RESULTS MET significantly inhibited inflammatory damage and oxidative stress responses in LPS-induced HTR-8/SVneo cells. Besides, MET could activate AMPK and then affect NF-κB/sFlt-1 and Nrf2/HO-1 signaling pathways in LPS-induced HTR-8/SVneo cells. Compound C (an AMPK inhibitor) significantly reversed MET's effects on LPS-stimulated HTR-8/SVneo cells. DISCUSSION MET attenuated inflammatory and oxidative stress of HTR-8/SVneo cells in PE by activating AMPK to regulate NF-κB/sFlt-1 and Nrf2/HO-1 signaling pathways, suggesting that MET was a potential therapeutic drug for PE.
Collapse
Affiliation(s)
- Lidan He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, Fujian, China.
| | - Xiuyan Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, Fujian, China
| | - Feng Zhan
- School of Electronic Information Engineering, Taiyuan University of Science and Technology, Taiyuan, 030024, Shanxi, China; College of Engineering, Fujian Jiangxia University, Fuzhou, 350108, Fujian, China
| | - Xuemei Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, Fujian, China
| | - Jianbo Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, Fujian, China.
| |
Collapse
|
184
|
Hurtado I, Bonacina E, Garcia-Manau P, Serrano B, Armengol-Alsina M, Mendoza M, Maiz N, Carreras E. Usefulness of angiogenic factors in prenatal counseling of late-onset fetal growth-restricted and small-for-gestational-age gestations: a prospective observational study. Arch Gynecol Obstet 2023; 308:1485-1495. [PMID: 36401095 DOI: 10.1007/s00404-022-06833-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/19/2022] [Indexed: 11/21/2022]
Abstract
OBJECTIVES To create a predictive model including biomarkers and evaluate its ability to predict adverse perinatal outcomes in late-onset small fetuses, ultimately helping to provide individualized counseling at the time of diagnosis. METHODS This was a prospective observational study, including singleton pregnancies with an estimated fetal weight (EFW) below the 10th percentile, at a gestational age between 32 + 0 and 36 + 6 weeks of gestation (WG). Variables recorded at diagnosis to predict adverse pregnancy outcomes were: soluble fms-like tyrosine-kinase-1 to placental growth factor ratio (sFlt-1/PlGF), fetal Doppler (umbilical artery and middle cerebral artery), uterine artery pulsatility index (UtAPI), EFW percentile, gestational age, and the presence of maternal risk factors for placental insufficiency. Logistic regression models were developed for the prediction of three co-primary outcomes: composite adverse perinatal outcomes (APO), and the need for elective delivery before 35 or 37 WG. RESULTS Sixty (52.2%) fetal growth restricted (FGR) and 55 (47.8%) small for gestational age (SGA) were enrolled. Thirteen (11.3%) women needed elective delivery before 35 WG and 27 (23.5%) women before 37 WG. At least one APO occurred in 43 (37.4%) pregnancies. The best marker in univariate analyses was the sFlt-1/PlGF ratio [AUC = 0.932 (95% CI, 0.864-0.999)]. The multivariate model including sFlt-1/PlGF showed a better predictive performance for APO than the multivariate model without sFlt-1/PlGF (P < 0.024). CONCLUSIONS sFlt-1/PlGF is a good predictor of APO at the time of late-onset FGR/SGA diagnosis. Our predictive models may be useful to provide early individualized prenatal counseling in this group of women. Further studies are needed to validate these preliminary findings in a larger cohort.
Collapse
Affiliation(s)
- Ivan Hurtado
- Department of Obstetrics, Maternal Fetal Medicine Unit, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Passeig de La Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Erika Bonacina
- Department of Obstetrics, Maternal Fetal Medicine Unit, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Passeig de La Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Pablo Garcia-Manau
- Department of Obstetrics, Maternal Fetal Medicine Unit, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Passeig de La Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Berta Serrano
- Department of Obstetrics, Maternal Fetal Medicine Unit, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Passeig de La Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Mireia Armengol-Alsina
- Department of Obstetrics, Maternal Fetal Medicine Unit, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Passeig de La Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Manel Mendoza
- Department of Obstetrics, Maternal Fetal Medicine Unit, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Passeig de La Vall d'Hebron, 119-129, 08035, Barcelona, Spain.
| | - Nerea Maiz
- Department of Obstetrics, Maternal Fetal Medicine Unit, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Passeig de La Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Elena Carreras
- Department of Obstetrics, Maternal Fetal Medicine Unit, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Passeig de La Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| |
Collapse
|
185
|
Sokratous N, Bednorz M, Wright A, Nicolaides KH, Kametas NA. Prediction using serum glycosylated fibronectin of imminent pre-eclampsia in women with new-onset hypertension. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2023; 62:653-659. [PMID: 37606310 DOI: 10.1002/uog.27458] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/03/2023] [Accepted: 08/11/2023] [Indexed: 08/23/2023]
Abstract
OBJECTIVE To compare the predictive performance for delivery with pre-eclampsia (PE) within 2 weeks after assessment in women with new-onset hypertension at 24-41 weeks' gestation between serum glycosylated fibronectin (GlyFn) concentration, serum placental growth factor (PlGF) concentration and soluble fms-like tyrosine kinase-1 (sFlt-1) to PlGF concentration ratio. METHODS This was a prospective observational study of 409 women with a singleton pregnancy presenting at 24-41 weeks' gestation with new-onset hypertension. The recommended cut-off for sFlt-1/PlGF ratio for the prediction of PE in the platform used in this study is 85; the appropriate cut-offs for GlyFn and PlGF were determined to achieve the same screen-positive rate as that of sFlt-1/PlGF ratio > 85. We then compared the predictive performance for delivery with PE within 2 weeks after presentation between GlyFn, PlGF and sFlt-1/PlGF, both overall and in subgroups according to gestational age at presentation. RESULTS Delivery with PE within 2 weeks occurred in 93 (22.7%) cases. The screen-positive rate for sFlt-1/PlGF ratio > 85 was 46.2%. The cut-off corresponding to a screen-positive rate of 46.2% was 75 pg/mL for PlGF and 510 µg/mL for GlyFn. The overall detection rate for delivery with PE within 2 weeks after presentation was 62.4% (95% CI, 51.7-72.2%) for GlyFn and sFlt-1/PlGF and 60.2% (95% CI, 49.5-70.2%) for PlGF. In all women who delivered with PE within 2 weeks after presentation at < 34 weeks' gestation and in about 60-70% of those presenting at < 38 weeks, GlyFn and sFlt-1/PlGF were increased and PlGF was reduced. However, the screen-positive rate for these tests was very high at about 45%. The predictive performance for delivery with PE within 2 weeks after presentation at ≥ 38 weeks' gestation was poorer for all three methods of screening, with detection rates of 47-63% at screen-positive rates of 40-50%. CONCLUSIONS In women with new-onset hypertension, the predictive performance for delivery with PE within 2 weeks after presentation for serum GlyFn is similar to that of PlGF and the sFlt-1/PlGF ratio, but GlyFn may be the preferred option because it is a rapid point-of-care test. However, the predictive performance for all tests is relatively poor. © 2023 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- N Sokratous
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - M Bednorz
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - A Wright
- Institute of Health Research, University of Exeter, Exeter, UK
| | - K H Nicolaides
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - N A Kametas
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| |
Collapse
|
186
|
Pérez-Gutiérrez L, Ferrara N. Biology and therapeutic targeting of vascular endothelial growth factor A. Nat Rev Mol Cell Biol 2023; 24:816-834. [PMID: 37491579 DOI: 10.1038/s41580-023-00631-w] [Citation(s) in RCA: 107] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2023] [Indexed: 07/27/2023]
Abstract
The formation of new blood vessels, called angiogenesis, is an essential pathophysiological process in which several families of regulators have been implicated. Among these, vascular endothelial growth factor A (VEGFA; also known as VEGF) and its two tyrosine kinase receptors, VEGFR1 and VEGFR2, represent a key signalling pathway mediating physiological angiogenesis and are also major therapeutic targets. VEGFA is a member of the gene family that includes VEGFB, VEGFC, VEGFD and placental growth factor (PLGF). Three decades after its initial isolation and cloning, VEGFA is arguably the most extensively investigated signalling system in angiogenesis. Although many mediators of angiogenesis have been identified, including members of the FGF family, angiopoietins, TGFβ and sphingosine 1-phosphate, all current FDA-approved anti-angiogenic drugs target the VEGF pathway. Anti-VEGF agents are widely used in oncology and, in combination with chemotherapy or immunotherapy, are now the standard of care in multiple malignancies. Anti-VEGF drugs have also revolutionized the treatment of neovascular eye disorders such as age-related macular degeneration and ischaemic retinal disorders. In this Review, we emphasize the molecular, structural and cellular basis of VEGFA action as well as recent findings illustrating unexpected interactions with other pathways and provocative reports on the role of VEGFA in regenerative medicine. We also discuss clinical and translational aspects of VEGFA. Given the crucial role that VEGFA plays in regulating angiogenesis in health and disease, this molecule is largely the focus of this Review.
Collapse
Affiliation(s)
- Lorena Pérez-Gutiérrez
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Napoleone Ferrara
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
- Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
187
|
Giardini V, Grilli L, Terzaghi A, Todyrenchuk L, Zavettieri C, Mazzoni G, Cozzolino S, Casati M, Vergani P, Locatelli A. sFlt-1 Levels as a Predicting Tool in Placental Dysfunction Complications in Multiple Pregnancies. Biomedicines 2023; 11:2917. [PMID: 38001918 PMCID: PMC10669317 DOI: 10.3390/biomedicines11112917] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND several studies have demonstrated that angiogenic markers can improve the clinical management of hypertensive disorders (HDs) and fetal growth restriction (FGR) in singleton pregnancies, but few studies have evaluated the performance of these tests in multiple pregnancies. Our aim was to investigate the role of soluble fms-like tyrosine kinase 1 (sFlt-1) in predicting adverse obstetric outcomes in hospitalized multiple pregnancies with HD (preeclampsia/gestational hypertension/uncontrolled chronic hypertension) and/or FGR in one or more fetuses. METHODS A retrospective analysis of multiple pregnancies with HD/FGR occurring after the 20th gestational week. Pregnant women were divided into two groups: women with high levels of sFlt-1 and those with low levels of sFlt-1. A value of sFlt-1 greater than or equal to 15,802 pg/mL was considered arbitrarily high, as it is equivalent to two times the 90th percentile expected in an uncomplicated full-term singleton pregnancy based on data from a prospective multicenter study (7901 pg/mL). RESULTS The cohort included 39 multiple pregnancies. There were no cases of birth <34 weeks, HELLP syndrome, ICU admission, and urgent cesarean sections for HD/FGR complications reported among women with low levels of sFlt-1. CONCLUSIONS A cut-off value of sFlt-1 ≥ 15,802 pg/mL could represent a valuable tool for predicting adverse obstetric outcomes in multiple pregnancies hospitalized for HD/FGR disorders, regardless of gestational age and chorionicity.
Collapse
Affiliation(s)
- Valentina Giardini
- Department of Obstetrics and Gynecology, IRCCS San Gerardo dei Tintori Foundation, University of Milano-Bicocca, 20900 Monza, Italy; (L.G.); (A.T.); (C.Z.); (A.L.)
| | - Leonora Grilli
- Department of Obstetrics and Gynecology, IRCCS San Gerardo dei Tintori Foundation, University of Milano-Bicocca, 20900 Monza, Italy; (L.G.); (A.T.); (C.Z.); (A.L.)
| | - Alessandra Terzaghi
- Department of Obstetrics and Gynecology, IRCCS San Gerardo dei Tintori Foundation, University of Milano-Bicocca, 20900 Monza, Italy; (L.G.); (A.T.); (C.Z.); (A.L.)
| | - Lyudmyla Todyrenchuk
- Department of Obstetrics and Gynecology, IRCCS San Gerardo dei Tintori Foundation, University of Milano-Bicocca, 20900 Monza, Italy; (L.G.); (A.T.); (C.Z.); (A.L.)
| | - Caterina Zavettieri
- Department of Obstetrics and Gynecology, IRCCS San Gerardo dei Tintori Foundation, University of Milano-Bicocca, 20900 Monza, Italy; (L.G.); (A.T.); (C.Z.); (A.L.)
| | - Giulia Mazzoni
- Department of Obstetrics and Gynecology, IRCCS San Gerardo dei Tintori Foundation, University of Milano-Bicocca, 20900 Monza, Italy; (L.G.); (A.T.); (C.Z.); (A.L.)
| | - Sabrina Cozzolino
- Department of Obstetrics and Gynecology, IRCCS San Gerardo dei Tintori Foundation, University of Milano-Bicocca, 20900 Monza, Italy; (L.G.); (A.T.); (C.Z.); (A.L.)
| | - Marco Casati
- Laboratory Medicine, IRCCS San Gerardo dei Tintori Foundation, University of Milano-Bicocca, 20900 Monza, Italy
| | - Patrizia Vergani
- Department of Obstetrics and Gynecology, IRCCS San Gerardo dei Tintori Foundation, University of Milano-Bicocca, 20900 Monza, Italy; (L.G.); (A.T.); (C.Z.); (A.L.)
| | - Anna Locatelli
- Department of Obstetrics and Gynecology, IRCCS San Gerardo dei Tintori Foundation, University of Milano-Bicocca, 20900 Monza, Italy; (L.G.); (A.T.); (C.Z.); (A.L.)
| |
Collapse
|
188
|
Takakura S, Tanaka H, Tamaishi Y, Enomoto N, Magawa S, Maki S, Nii M, Tanaka K, Toriyabe K, Kondo E, Ikeda T. The role of periodic measurement of sFlt-1 and PlGF in predicting the remaining pregnancy duration in hypertensive disorders of pregnancy. Pregnancy Hypertens 2023; 34:104-109. [PMID: 39492240 DOI: 10.1016/j.preghy.2023.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/11/2023] [Accepted: 10/06/2023] [Indexed: 11/05/2024]
Abstract
OBJECTIVES This study aimed to explore whether one-time or periodic measurement of soluble fms-like tyrosine kinase-1 (sFlt-1) and placental growth factor (PlGF) could contribute to the prediction of the remaining pregnancy duration in hypertensive disorders of pregnancy. STUDY DESIGN In this retrospective study, we enrolled pregnant women with singleton pregnancies who were admitted to our hospital due to a new rise in blood pressure at or after 20 weeks of gestation or because of worsening chronic hypertension (CH). MAIN OUTCOME MEASURES The concentrations of sFlt-1 and PlGF, as well as the sFlt-1/PlGF ratio, were measured only on admission or on admission and every week until delivery. The effect of these concentrations was correlated with the remaining pregnancy duration. RESULTS In this study, we enrolled 32 pregnant women: 13 were diagnosed with preeclampsia (PE), 9 with gestational hypertension (GH), and 10 with CH on admission. In the PE group, the concentration of sFlt-1 on admission had a significant negative correlation with the remaining pregnancy duration (R = -0.61, P = 0.03). In three women with PE, the concentrations were measured periodically, with increasing sFlt-1 concentrations. Among the pregnant women with CH on admission, two developed superimposed PE and their measured concentrations of sFlt-1 periodically increased. CONCLUSIONS Only the concentration of sFlt-1 on admission in the PE group was associated with the remaining pregnancy duration. The concentrations of sFlt-1 in PE cases periodically measured increased towards delivery. Pregnant women with CH could develop sPE if the sFlt-1 value measured periodically increase.
Collapse
Affiliation(s)
- Sho Takakura
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Tsu, Mie, Japan.
| | - Hiroaki Tanaka
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Tsu, Mie, Japan
| | - Yuya Tamaishi
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Tsu, Mie, Japan
| | - Naosuke Enomoto
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Tsu, Mie, Japan
| | - Shoichi Magawa
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Tsu, Mie, Japan
| | - Shintaro Maki
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Tsu, Mie, Japan
| | - Masafumi Nii
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Tsu, Mie, Japan
| | - Kayo Tanaka
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Tsu, Mie, Japan
| | - Kuniaki Toriyabe
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Tsu, Mie, Japan
| | - Eiji Kondo
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Tsu, Mie, Japan
| | - Tomoaki Ikeda
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Tsu, Mie, Japan
| |
Collapse
|
189
|
Yin Y, Xie S, Xu Q, Liao L, Chen H, Zhou R. Circulating chemerin levels in preeclampsia: a systematic review and meta-analysis. Lipids Health Dis 2023; 22:179. [PMID: 37864182 PMCID: PMC10588206 DOI: 10.1186/s12944-023-01941-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/13/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND Preeclampsia (PE) is a new-onset pregnancy-specific disorder with a high prevalence that leads to over 70 000 maternal and 500 000 foetal fatalities worldwide each year. The level of chemerin, a newly identified adipokine, is increased in diabetic and obese patients. Currently, there are several studies describing the relationship between maternal circulating chemerin levels and PE. Therefore, this study aimed to assess their association in pooled samples. METHODS Four databases were systematically searched to identify potential studies that reported circulating chemerin levels in PE and normal pregnancy groups. Standardized mean differences (SMDs), 95% confidence intervals (CIs), and 95% prediction intervals (PIs) were calculated using a random-effects meta-analysis. The probability of heterogeneity was also investigated by sensitivity analysis, subgroup analysis, and meta-regression. RESULTS Thirteen studies in 11 articles with a total of 860 PE patients and 1309 women with normal pregnancies met the inclusion criteria. The results of the meta-analysis revealed that circulating chemerin, which levels in PE patients were considerably higher than those in controls (SMD = 1.39, 95% CI: 1.02, 1.77, 95% PI: -0.07, 2.86). Moreover, sensitivity analysis determined that the outcomes of the overall pooled results were not affected after the elimination of any study. Notably, subgroup analysis demonstrated a similar expression pattern irrespective of geographic location, severity, timing of sampling, and sample size. Last, there were no factors that significantly impacted the overall estimate, according to meta-regression. CONCLUSIONS This meta-analysis is the first to assess circulating chemerin levels in PE patients. The findings indicate that circulating chemerin levels may be a potential marker to diagnose PE.
Collapse
Affiliation(s)
- Yangxue Yin
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, No. 20, section 3, Renmin South Road, Wuhou District, Chengdu, Sichuan, 610041, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Shuangshuang Xie
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, No. 20, section 3, Renmin South Road, Wuhou District, Chengdu, Sichuan, 610041, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Qin Xu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, No. 20, section 3, Renmin South Road, Wuhou District, Chengdu, Sichuan, 610041, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Lingyun Liao
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, No. 20, section 3, Renmin South Road, Wuhou District, Chengdu, Sichuan, 610041, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Hongqin Chen
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, No. 20, section 3, Renmin South Road, Wuhou District, Chengdu, Sichuan, 610041, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Rong Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, No. 20, section 3, Renmin South Road, Wuhou District, Chengdu, Sichuan, 610041, China.
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China.
| |
Collapse
|
190
|
Rimboeck J, Gruber M, Weigl M, Huber P, Lunz D, Petermichl W. Obesity Correlates with Chronic Inflammation of the Innate Immune System in Preeclampsia and HELLP Syndrome during Pregnancy. Biomedicines 2023; 11:2851. [PMID: 37893224 PMCID: PMC10604126 DOI: 10.3390/biomedicines11102851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/14/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
HELLP syndrome is characterized by hemolysis, elevated liver enzymes, and a low platelet count and poses an increased risk to the pregnant woman and the unborn child. Individual risk factors such as obesity may alter immunocompetence and influence the course of preeclampsia (PE) or HELLP syndrome. Blood samples were collected from 21 pregnant women (7 healthy, 6 with PE, and 8 with HELLP syndrome) and polymorphonuclear neutrophils (PMNs) were subsequently isolated. Production of radical oxygen species (ROS), cell movement, and NETosis were assessed by live-cell imaging. Surface protein expression and oxidative burst were analyzed by flow cytometry. PE and HELLP patients had significantly higher BMI compared to the healthy control group. Depending on the expression of CD11b, CD62L, and CD66b on PMNs, a surface protein activation sum scale (SPASS) was calculated. PMNs from patients with high SPASS values showed prolonged and more targeted migration with delayed ROS production and NETosis. Obesity is associated with a chronic inflammatory state, which in combination with immunological triggers during pregnancy could modulate PMN functions. Pregnant women with higher BMI tend to have higher SPASS values, indicating activation of the innate immune system that could co-trigger PE or HELLP syndrome.
Collapse
Affiliation(s)
- Julia Rimboeck
- Department of Anesthesiology, University Hospital of Regensburg, 93042 Regensburg, Germany
| | - Michael Gruber
- Department of Anesthesiology, University Hospital of Regensburg, 93042 Regensburg, Germany
| | - Marco Weigl
- University Department of Obstetrics and Gynecology at the Hospital St. Hedwig of the Order of St. John, University of Regensburg, 93049 Regensburg, Germany
| | - Pia Huber
- Department of Anesthesiology, University Hospital of Regensburg, 93042 Regensburg, Germany
| | - Dirk Lunz
- Department of Anesthesiology, University Hospital of Regensburg, 93042 Regensburg, Germany
| | - Walter Petermichl
- Department of Anesthesiology, University Hospital of Regensburg, 93042 Regensburg, Germany
| |
Collapse
|
191
|
Admati I, Skarbianskis N, Hochgerner H, Ophir O, Weiner Z, Yagel S, Solt I, Zeisel A. Two distinct molecular faces of preeclampsia revealed by single-cell transcriptomics. MED 2023; 4:687-709.e7. [PMID: 37572658 DOI: 10.1016/j.medj.2023.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/04/2023] [Accepted: 07/14/2023] [Indexed: 08/14/2023]
Abstract
INTRODUCTION Preeclampsia is a multisystemic, pregnancy-specific disorder united by new-onset hypertension but with considerable variation in clinical manifestation, onset, and severity. For symptoms to regress, delivery of the placenta is required. For symptoms to regress, delivery of the placenta is required, making the placenta central to preeclampsia pathophysiology. To dissect which placental functions were impacted in two forms of preeclampsia, we studied molecular changes across the cell types of the placenta. METHODS We performed a transcriptomic survey of single-cells and single-nuclei on cases of early- and late-onset preeclampsia with gestation-matched controls. FINDINGS Our data revealed massive dysregulation of gene expression in all cell classes that was almost exclusive to early preeclampsia. For example, an important known receptor/ligand imbalance hallmarking angiogenic disfunction, sFLT1/placental growth factor (PGF), was reflected in striking, cell-autonomous dysregulation of FLT1 and PGF transcription in the syncytium in early preeclampsia only. Stromal cells and vasculature echoed an inflamed, stressed, anti-angiogenic environment. Finally, the placental immune niche set the tone for inflammation in early but not late preeclampsia. Here, fetal-origin Hofbauer and maternal-origin TREM2 macrophages were revealed as surprising main actors, while local cells of the adaptive immune system were largely unaffected. Late preeclampsia showed minimal cellular impact on the placenta. CONCLUSIONS Our survey provides systematic molecular evidence for two distinct diseases. We resolved systematic molecular dysregulation to individual cell types with strong implications for definition, early detection, diagnosis, and treatment. FUNDING Funded by the Preeclampsia Foundation through the Peter Joseph Pappas Research Grant.
Collapse
Affiliation(s)
- Inbal Admati
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Niv Skarbianskis
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Hannah Hochgerner
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Osnat Ophir
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Zeev Weiner
- Department of Obstetrics and Gynecology, Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Simcha Yagel
- Division of Obstetrics and Gynecology Hadassah, Hebrew University Medical Centers, Jerusalem, Israel
| | - Ido Solt
- Department of Obstetrics and Gynecology, Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel.
| | - Amit Zeisel
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
192
|
Pané A, Milad C, Santana-Domínguez M, Baños N, Borras-Novell C, Espinosa G, Magnano L, Nomdedeu M, Moreno-Lozano PJ, Cofan F, Placeres M, Fernández RM, García-Villoria J, Garrabou G, Vinagre I, Tanner LM, Montserrat-Carbonell C, Forga-Visa MDT. Lysinuric Protein Intolerance and Its Nutritional and Multisystemic Challenges in Pregnancy: A Case Report and Literature Review. J Clin Med 2023; 12:6405. [PMID: 37835050 PMCID: PMC10573933 DOI: 10.3390/jcm12196405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/01/2023] [Accepted: 10/07/2023] [Indexed: 10/15/2023] Open
Abstract
Lysinuric protein intolerance (LPI) is a rare inborn error of metabolism (IEM), classified as an inherited aminoaciduria, caused by mutations in the SLC7A7 gene, leading to a defective cationic amino acid transport. The metabolic adaptations to the demands of pregnancy and delivery cause significant physiological stress, so those patients affected by IEM are at greater risk of decompensation. A 28-year-old woman with LPI had experienced 3 early miscarriages. While pregnancy was finally achieved, diverse nutritional and medical challenges emerged (food aversion, intrauterine growth restriction, bleeding risk, and preeclampsia suspicion), which put both the mother and the fetus at risk. Moreover, the patient requested a natural childbirth (epidural-free, delayed cord clamping). Although the existence of multiple safety concerns rejected this approach at first, the application of novel strategies made a successful delivery possible. This case reinforces that the woman's wish for a non-medicated, low-intervention natural birth should not be automatically discouraged because of an underlying complex metabolic condition. Achieving a successful pregnancy is conceivable thanks to the cooperation of interdisciplinary teams, but it is still important to consider the risks beforehand in order to be prepared for possible additional complications.
Collapse
Affiliation(s)
- Adriana Pané
- Endocrinology and Nutrition Department, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
- Adult Inborn Errors of Metabolism Unit, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Camila Milad
- Endocrinology and Nutrition Department, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
| | - Marta Santana-Domínguez
- Neonatology Department, BCNatal (Barcelona Center for Fetal and Neonatal Medicine), Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
| | - Núria Baños
- Neonatology Department, BCNatal (Barcelona Center for Fetal and Neonatal Medicine), Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
- Fundació Clínic per la Recerca Biomèdica (FCR), Institut d’Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Cristina Borras-Novell
- Neonatology Department, BCNatal (Barcelona Center for Fetal and Neonatal Medicine), Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
| | - Gerard Espinosa
- Fundació Clínic per la Recerca Biomèdica (FCR), Institut d’Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Autoimmune Diseases Unit, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
| | - Laura Magnano
- Fundació Clínic per la Recerca Biomèdica (FCR), Institut d’Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Department of Hematology, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
| | - Meritxell Nomdedeu
- Department of Hematology, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
| | - Pedro Juan Moreno-Lozano
- Adult Inborn Errors of Metabolism Unit, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
- Internal Medicine Department, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
- Asociación Española para el Estudio de los Errores Congénitos del Metabolismo (AECOM), 28221 Majadahonda, Spain
| | - Frederic Cofan
- Renal Transplantation and Nephrology Department, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
| | - Mercè Placeres
- Adult Inborn Errors of Metabolism Unit, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
- Pharmacy Department, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
| | - Rosa Maria Fernández
- Biochemistry and Molecular Genetics Department, Biomedical Diagnostic Center, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
| | - Judit García-Villoria
- Adult Inborn Errors of Metabolism Unit, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
- Asociación Española para el Estudio de los Errores Congénitos del Metabolismo (AECOM), 28221 Majadahonda, Spain
- Biochemistry and Molecular Genetics Department, Biomedical Diagnostic Center, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Glòria Garrabou
- Adult Inborn Errors of Metabolism Unit, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
- Biochemistry and Molecular Genetics Department, Biomedical Diagnostic Center, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Inherited Metabolic Diseases and Muscle Disorders Laboratory, FCRB-IDIBAPS and Faculty of Medicine and Heath Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Irene Vinagre
- Endocrinology and Nutrition Department, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
- Fundació Clínic per la Recerca Biomèdica (FCR), Institut d’Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Laura M. Tanner
- Fetomaternal Medical Center and Department of Clinical Genetics, Helsinki University Hospital, Department of Medical and Clinical Genetics, University of Helsinki, 00251 Helsinki, Finland
| | - Cristina Montserrat-Carbonell
- Endocrinology and Nutrition Department, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
- Adult Inborn Errors of Metabolism Unit, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
- Asociación Española para el Estudio de los Errores Congénitos del Metabolismo (AECOM), 28221 Majadahonda, Spain
| | - Maria de Talló Forga-Visa
- Endocrinology and Nutrition Department, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
- Adult Inborn Errors of Metabolism Unit, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain
- Asociación Española para el Estudio de los Errores Congénitos del Metabolismo (AECOM), 28221 Majadahonda, Spain
| |
Collapse
|
193
|
Velegrakis A, Kouvidi E, Fragkiadaki P, Sifakis S. Predictive value of the sFlt‑1/PlGF ratio in women with suspected preeclampsia: An update (Review). Int J Mol Med 2023; 52:89. [PMID: 37594116 PMCID: PMC10500221 DOI: 10.3892/ijmm.2023.5292] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/13/2023] [Indexed: 08/19/2023] Open
Abstract
Preeclampsia (PE) is a major complication of pregnancy with an incidence rate of 2‑8% and is a leading cause of maternal mortality and morbidity. The various consequences of severe preeclampsia for the fetus, neonate and child include intrauterine growth retardation (IUGR), fetal hypoxia, oligohydramnios, intrauterine fetal demise, increased perinatal mortality and morbidity, neurodevelopmental disorders and even irreversible brain damage (cerebral palsy). A number of studies have demonstrated that differences in maternal serum concentrations of angiogenic factors between preeclampsia and normotensive pregnancies can be used as biomarkers, either alone or in combination with other markers, to predict the development of PE. The presence in the maternal circulation of two proteins of placental origin, placental growth factor (PlGF) and soluble fms‑like tyrosine kinase 1 (sFlt‑1), has been shown to be of clinical value, as the sFlt‑1/PlGF ratio appears to be the optimal predictive tool for the development of PE. The measurement of their concentration in maternal serum in screening models, serves as predictive marker for the development of PE or IUGR later in gestation. However, further research is required to improve its clinical applicability and provide guidelines for its use worldwide to achieve more consistent clinical management of women with PE.
Collapse
Affiliation(s)
- Alexandros Velegrakis
- Department of Obstetrics and Gynecology, University Hospital of Heraklion, 71500 Heraklion, Greece
| | - Elisavet Kouvidi
- Genesis Genoma Lab, Genetic Diagnosis, Clinical Genetics and Research, 15232 Athens, Greece
| | - Persefoni Fragkiadaki
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece
| | | |
Collapse
|
194
|
Bajpai D, Popa C, Verma P, Dumanski S, Shah S. Evaluation and Management of Hypertensive Disorders of Pregnancy. KIDNEY360 2023; 4:1512-1525. [PMID: 37526641 PMCID: PMC10617800 DOI: 10.34067/kid.0000000000000228] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 07/24/2023] [Indexed: 08/02/2023]
Abstract
Hypertensive disorders of pregnancy complicate up to 10% of pregnancies and remain the major cause of maternal and neonatal morbidity and mortality. Hypertensive disorders of pregnancy can be classified into four groups depending on the onset of hypertension and the presence of target organ involvement: chronic hypertension, preeclampsia, gestational hypertension, and superimposed preeclampsia on chronic hypertension. Hypertension during pregnancy is associated with a higher risk of cardiovascular disease and kidney failure. Early diagnosis and proper treatment for pregnant women with hypertension remain a priority since this leads to improved maternal and fetal outcomes. Labetalol, nifedipine, methyldopa, and hydralazine are the preferred medications to treat hypertension during pregnancy. In this comprehensive review, we discuss the diagnostic criteria, evaluation, and management of pregnant women with hypertension.
Collapse
Affiliation(s)
- Divya Bajpai
- Department of Nephrology, Seth G.S.M.C & K.E.M. Hospital, Mumbai, India
| | - Cristina Popa
- Department of Internal Medicine - Nephrology, University of Medicine and Pharmacy “Grigore T Popa”, Iasi, Romania
| | - Prasoon Verma
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Sandi Dumanski
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Libin Cardiovascular Institute, Calgary, Alberta, Canada
- Alberta Kidney Disease Network, Calgary, Alberta, Canada
| | - Silvi Shah
- Division of Nephrology and Hypertension, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
195
|
Rao A, Shinde U, Das DK, Balasinor N, Madan T. Early prediction of pre-eclampsia using circulating placental exosomes: Newer insights. Indian J Med Res 2023; 158:385-396. [PMID: 37987999 DOI: 10.4103/ijmr.ijmr_2143_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Indexed: 11/22/2023] Open
Abstract
Pre-eclampsia (PE), a multifactorial de novo hypertensive pregnancy disorder, is one of the leading causes of foeto-maternal morbidity and mortality. Currently, antihypertensive drugs are the first-line therapy for PE and evidence suggests that low-dose aspirin initiated early in high risk pregnancies may reduce the risk of development or severity of PE. However, an early prediction of this disorder remains an unmet clinical challenge. Several potential serum biomarkers associated with maternal immunoregulation and placental angiogenesis have been evaluated but are ineffective and inconsistent for early prediction. Although placental biomarkers would be more specific and sensitive in predicting the risk of PE, accessing the placenta during pregnancy is not feasible. Circulating placental exosomes (pEXO), originating from foeto-maternal interface, are being evaluated as the placenta's surrogate and the best source of non-invasive placental biomarkers. pEXO appear in the maternal circulation starting from six weeks of gestation and its dynamic biological cargo across pregnancy is associated with successful pregnancy outcomes. Therefore, monitoring changes in pEXO expression profiles could provide new insights into the prediction, diagnosis and treatment of PE. This narrative review comprehensively summarizes the available literature on the candidate predictive circulating biomarkers evaluated for PE to date. In particular, the review elucidates the current knowledge of distinct molecular signatures emanating from pEXO in pre-eclamptic women to support the discovery of novel early predictive biomarkers for effective intervention and management of the disease.
Collapse
Affiliation(s)
- Aishwarya Rao
- Department of Innate Immunity, ICMR-National Institute for Research in Reproductive & Child Health, Mumbai, Maharashtra, India
| | - Uma Shinde
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive & Child Health, Mumbai, Maharashtra, India
| | - Dhanjit Kumar Das
- Department of Stem Cell Biology, ICMR-National Institute for Research in Reproductive & Child Health, Mumbai, Maharashtra, India
| | - Nafisa Balasinor
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive & Child Health, Mumbai, Maharashtra, India
| | - Taruna Madan
- Department of Innate Immunity, ICMR-National Institute for Research in Reproductive & Child Health, Mumbai, Maharashtra, India
| |
Collapse
|
196
|
Bisson C, Rajan RS, Rana S. Angiogenic Biomarkers for Preeclampsia in Clinical Setting: A New Era. Hypertension 2023; 80:2029-2032. [PMID: 37729636 DOI: 10.1161/hypertensionaha.123.21686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Affiliation(s)
- Courtney Bisson
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago Medicine, IL (C.B., S.R.)
| | - Revathi S Rajan
- Department of Maternal Fetal Medicine, Mirror Health, Bengaluru, India (R.S.R.)
| | - Sarosh Rana
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago Medicine, IL (C.B., S.R.)
| |
Collapse
|
197
|
De Oliveira L, Roberts JM, Jeyabalan A, Blount K, Redman CW, Poston L, Seed PT, Chappell LC, Dias MAB. PREPARE: A Stepped-Wedge Cluster-Randomized Trial to Evaluate Whether Risk Stratification Can Reduce Preterm Deliveries Among Patients With Suspected or Confirmed Preterm Preeclampsia. Hypertension 2023; 80:2017-2028. [PMID: 37431663 PMCID: PMC10510842 DOI: 10.1161/hypertensionaha.122.20361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 06/19/2023] [Indexed: 07/12/2023]
Abstract
BACKGROUND Early delivery in preterm preeclampsia may reduce the risks for the patient, but consequences of prematurity may be substantial for the baby. This trial evaluated whether the implementation of a risk stratification model could safely reduce prematurity. METHODS This was a stepped-wedge cluster-randomized trial in seven clusters. Patients presenting with suspected or confirmed preeclampsia between 20+0 and 36+6 gestational weeks were considered eligible. At the start of the trial, all centers were allocated in the preintervention phase, and patients enrolled in this phase were managed according to local treatment guidance. Subsequently, every 4 months, 1 randomly allocated cluster transitioned to the intervention. Patients enrolled in the intervention phase had sFlt-1 (soluble fms-like tyrosine kinase-1)/PlGF (placental growth factor) ratio and preeclampsia integrated estimate of risk assessments performed. If sFlt-1/PlGF ≤38 and preeclampsia integrated estimate of risk <10%, patients were considered low risk and clinicians received recommendations to defer delivery. If sFlt-1/PlGF >38 and preeclampsia integrated estimate of risk ≥10%, patients were considered not low risk, and clinicians received recommendations to increase surveillance. The primary outcome was the proportion of patients with preterm preeclampsia delivered prematurely out of total deliveries. RESULTS Between March 25, 2017 and December 24, 2019, 586 and 563 patients were analyzed in the intervention and usual care groups, respectively. The event rate was 1.09% in the intervention group, and 1.37% in the usual care group. After prespecified adjustments for variation between and within clusters over time, the adjusted risk ratio was 1.45 ([95% CI, 1.04-2.02]; P=0.029), indicating a higher risk of preterm deliveries in the intervention group. Post hoc analysis including calculation of risk differences did not show evidence of statistical differences. Abnormal sFlt-1/PlGF was associated with a higher rate of identifying preeclampsia with severe features. CONCLUSIONS The introduction of an intervention based on biomarkers and clinical factors for risk stratification did not lead to reductions in preterm deliveries. Further training on the interpretation of disease severity in preeclampsia and the development of additional risk stratification is needed before adoption into clinical practice. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT03073317.
Collapse
Affiliation(s)
- Leandro De Oliveira
- Botucatu Medical School, Obstetrics Department, Botucatu Sao Paulo State University, SP, Brazil (L.D.O.)
| | - James M. Roberts
- Magee-Womens Research Institute Department of Obstetrics and Gynecology, Epidemiology and Clinical and Translational Research, Pittsburgh, PA (J.M.R., A.J., K.B.)
| | - Arundhathi Jeyabalan
- Magee-Womens Research Institute Department of Obstetrics and Gynecology, Epidemiology and Clinical and Translational Research, Pittsburgh, PA (J.M.R., A.J., K.B.)
| | - Kasey Blount
- Magee-Womens Research Institute Department of Obstetrics and Gynecology, Epidemiology and Clinical and Translational Research, Pittsburgh, PA (J.M.R., A.J., K.B.)
| | - Christopher W. Redman
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, United Kingdom (C.W.R.)
| | - Lucilla Poston
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College of London, United Kingdom (L.P., P.T.S., L.C.C.)
| | - Paul T. Seed
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College of London, United Kingdom (L.P., P.T.S., L.C.C.)
| | - Lucy C. Chappell
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College of London, United Kingdom (L.P., P.T.S., L.C.C.)
| | | |
Collapse
|
198
|
Lara B, Sassot M, Calo G, Paparini D, Gliosca L, Chaufan G, Loureiro I, Vota D, Ramhorst R, Pérez Leirós C, Hauk V. Extracellular Vesicles of Porphyromonas gingivalis Disrupt Trophoblast Cell Interaction with Vascular and Immune Cells in an In Vitro Model of Early Placentation. Life (Basel) 2023; 13:1971. [PMID: 37895353 PMCID: PMC10608595 DOI: 10.3390/life13101971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Extracellular vesicles released by the primary pathogen of periodontal disease Porphyromonas gingivalis (Pg), referred to as outer membrane vesicles (OMVs), have been associated with the pathogenesis of systemic diseases like cardiovascular disease, rheumatoid arthritis, and Alzheimer's disease. A pathogenic role for Pg by disrupting placental homeostasis was proposed in the association between periodontal disease and adverse pregnancy outcomes. On the basis that trophoblast-derived factors modulate endothelial and immune cell profiles in normal pregnancy and the scarce presence of Pg in placenta, we hypothesized that OMVs from Pg affect trophoblast cell phenotype, impairing trophoblast-endothelium and trophoblast-neutrophil interactions. By means of in vitro designs with first-trimester human trophoblast cells, endothelial cells, and freshly isolated neutrophils, we showed that Pg OMVs are internalized by trophoblast cells and modulate the activity and expression of functional markers. Trophoblast cells primed with Pg OMVs enhanced neutrophil chemoattraction and lost their anti-inflammatory effect. In addition, reduced migration with enhanced adhesion of monocytes was found in endothelial cells upon incubation with the media from trophoblast cells pretreated with Pg OMVs. Taken together, the results support a pathogenic role of Pg OMVs at early stages of pregnancy and placentation through disruption of trophoblast contribution to vascular transformation and immune homeostasis maintenance.
Collapse
Affiliation(s)
- Brenda Lara
- Universidad de Buenos Aires—CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina; (B.L.); (M.S.); (G.C.); (D.P.); (L.G.); (G.C.); (I.L.); (D.V.); (R.R.)
| | - Matías Sassot
- Universidad de Buenos Aires—CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina; (B.L.); (M.S.); (G.C.); (D.P.); (L.G.); (G.C.); (I.L.); (D.V.); (R.R.)
| | - Guillermina Calo
- Universidad de Buenos Aires—CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina; (B.L.); (M.S.); (G.C.); (D.P.); (L.G.); (G.C.); (I.L.); (D.V.); (R.R.)
| | - Daniel Paparini
- Universidad de Buenos Aires—CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina; (B.L.); (M.S.); (G.C.); (D.P.); (L.G.); (G.C.); (I.L.); (D.V.); (R.R.)
| | - Laura Gliosca
- Universidad de Buenos Aires—CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina; (B.L.); (M.S.); (G.C.); (D.P.); (L.G.); (G.C.); (I.L.); (D.V.); (R.R.)
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Microbiología, Buenos Aires C1122AAH, Argentina
| | - Gabriela Chaufan
- Universidad de Buenos Aires—CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina; (B.L.); (M.S.); (G.C.); (D.P.); (L.G.); (G.C.); (I.L.); (D.V.); (R.R.)
| | - Iñaki Loureiro
- Universidad de Buenos Aires—CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina; (B.L.); (M.S.); (G.C.); (D.P.); (L.G.); (G.C.); (I.L.); (D.V.); (R.R.)
| | - Daiana Vota
- Universidad de Buenos Aires—CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina; (B.L.); (M.S.); (G.C.); (D.P.); (L.G.); (G.C.); (I.L.); (D.V.); (R.R.)
| | - Rosanna Ramhorst
- Universidad de Buenos Aires—CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina; (B.L.); (M.S.); (G.C.); (D.P.); (L.G.); (G.C.); (I.L.); (D.V.); (R.R.)
| | - Claudia Pérez Leirós
- Universidad de Buenos Aires—CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina; (B.L.); (M.S.); (G.C.); (D.P.); (L.G.); (G.C.); (I.L.); (D.V.); (R.R.)
| | - Vanesa Hauk
- Universidad de Buenos Aires—CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina; (B.L.); (M.S.); (G.C.); (D.P.); (L.G.); (G.C.); (I.L.); (D.V.); (R.R.)
| |
Collapse
|
199
|
Mahadevan A, Tipler A, Jones H. Shared developmental pathways of the placenta and fetal heart. Placenta 2023; 141:35-42. [PMID: 36604258 DOI: 10.1016/j.placenta.2022.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022]
Abstract
Congenital heart defects (CHD) remain the most common class of birth defect worldwide, affecting 1 in every 110 live births. A host of clinical and morphological indicators of placental dysfunction are observed in pregnancies complicated by fetal CHD and, with the recent emergence of single-cell sequencing capabilities, the molecular and physiological associations between the embryonic heart and developing placenta are increasingly evident. In CHD pregnancies, a hostile intrauterine environment may negatively influence and alter fetal development. Placental maldevelopment and dysfunction creates this hostile in-utero environment and may manifest in the development of various subtypes of CHD, with downstream perfusion and flow-related alterations leading to yet further disruption in placental structure and function. The adverse in-utero environment of CHD-complicated pregnancies is well studied, however the specific etiological role that the placenta plays in CHD development remains unclear. Many mouse and rat models have been used to characterize the relationship between CHD and placental dysfunction, but these paradigms present substantial limitations in the assessment of both the heart and placenta. Improvements in non-invasive placental assessment can mitigate these limitations and drive human-specific investigation in relation to fetal and placental development. Here, we review the clinical, structural, and molecular relationships between CHD and placental dysfunction, the CHD subtype-dependence of these changes, and the future of Placenta-Heart axis modeling and investigation.
Collapse
Affiliation(s)
- Aditya Mahadevan
- Physiology and Aging, University of Florida College of Medicine, USA; Center for Research in Perinatal Outcomes, University of Florida, USA
| | - Alyssa Tipler
- Physiology and Aging, University of Florida College of Medicine, USA; Center for Research in Perinatal Outcomes, University of Florida, USA
| | - Helen Jones
- Physiology and Aging, University of Florida College of Medicine, USA; Center for Research in Perinatal Outcomes, University of Florida, USA.
| |
Collapse
|
200
|
Abstract
Preeclampsia is a multisystem hypertensive disorder and one of the leading causes of maternal and fetal morbidity and mortality. The clinical hallmarks such as hypertension and proteinuria, and additional laboratory tests currently available including liver enzyme testing, are neither specific nor sufficiently sensitive. Therefore, biomarkers for timely and accurate identification of patients at risk of developing preeclampsia are extremely valuable to improve patient outcomes and safety. In this chapter, we will first discuss the clinical characteristics of preeclampsia and current evidence of the role of angiogenic factors, such as placental growth factor (PlGF) and soluble FMS like tyrosine kinase 1 (sFlt-1) in the pathogenesis of preeclampsia. Second, we will review the clinical practice guidelines for preeclampsia diagnostic criteria and their recommendations on laboratory testing. Third, we will review the currently available PlGF and sFlt-1 assays in terms of their methodologies, analytical performance, and clinical diagnostic values. Finally, we will discuss the future research needs from both an analytical and clinical perspective.
Collapse
Affiliation(s)
| | - Victoria Higgins
- DynaLIFE Medical Labs, Edmonton, AB, Canada; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Annie Ren
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Samantha Logan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Paul M Yip
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Precision Diagnostics and Therapeutics Program (Laboratory Medicine), Sunnybrook Health Sciences Center, Toronto, ON, Canada; Sunnybrook Research Institute, Toronto, ON, Canada
| | - Lei Fu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Precision Diagnostics and Therapeutics Program (Laboratory Medicine), Sunnybrook Health Sciences Center, Toronto, ON, Canada; Sunnybrook Research Institute, Toronto, ON, Canada.
| |
Collapse
|