1
|
Silasi M, Azzi M, Potchileev S, Burns L, Rana S. Placental Biomarker Testing for Evaluation of Suspected Preeclampsia. Clin Chem 2025; 71:548-558. [PMID: 40129181 DOI: 10.1093/clinchem/hvaf024] [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: 07/07/2024] [Accepted: 01/21/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND Worldwide, hypertensive disorders of pregnancy (HDP) are a leading cause of maternal and neonatal morbidity and mortality (Semin Perinatol 2009;33:130-7). This is especially true in the United States where preeclampsia is a leading cause of premature births (Hypertens Pregnancy 2016;35:510-9 and Lancet 2008;371:164-75). Moreover, this disorder is costly due to the financial burden of the health services needed to care for mothers with preeclampsia and their very often preterm infants (Am J Obstet Gynecol 2017;217:235-6). Recently, placental biomarkers have been shown to aid in assessment of the risk of severe preeclampsia. In 2023, the FDA approved the use of soluble feline McDonough sarcoma (fms)-like tyrosine kinase-1 to placental growth factor ratio (sFlt-1/PlGF) as an additional tool for preeclampsia risk assessment between 23 and 35 weeks' gestation in high-risk patients in the United States. Use of these biomarkers will improve maternal and fetal/neonatal outcomes and may assist in decreasing the healthcare burden of these patients by adding to risk assessment and the current diagnosis and management of pregnancies with HDP. CONTENT The pathophysiology of preeclampsia stems from abnormal placentation that results in an imbalance of pro- and antiangiogenic factors leading to endothelial and vascular dysfunction and the clinical syndrome of preeclampsia (J Clin Invest 2003;111:649-58). The role of the sFlt-1/PlGF in the prediction of progression to preeclampsia has been demonstrated in multiple studies. SUMMARY The goal of this review is to demonstrate the role of placental biomarkers (sFlt-1 and PlGF) in the pathophysiology of preeclampsia, with an emphasis on clinical applications and cost-effectiveness in the United States, using real-world applications as examples.
Collapse
Affiliation(s)
- Michelle Silasi
- Division of Maternal Fetal Medicine, Department of Obstetrics & Gynecology, Mercy Hospital St. Louis, St. Louis, MO, United States
| | - Marly Azzi
- Division of Maternal Fetal Medicine, Department of Obstetrics & Gynecology, University of Chicago Medical Center, Chicago, IL, United States
| | - Sanela Potchileev
- Division of Maternal Fetal Medicine, Department of Obstetrics & Gynecology, University of Chicago Medical Center, Chicago, IL, United States
| | - Luke Burns
- Division of Maternal Fetal Medicine, Department of Obstetrics & Gynecology, University of Chicago Medical Center, Chicago, IL, United States
| | - Sarosh Rana
- Division of Maternal Fetal Medicine, Department of Obstetrics & Gynecology, University of Chicago Medical Center, Chicago, IL, United States
| |
Collapse
|
2
|
Botha SM, Bartho LA, Hartmann S, Cannon P, Nguyen A, Nguyen TV, Pritchard N, Dechend R, Nonn O, Tong S, Kaitu'u-Lino TJ. Cystatin 6 (CST6) and Legumain (LGMN) are potential mediators in the pathogenesis of preeclampsia. Sci Rep 2025; 15:12945. [PMID: 40234537 PMCID: PMC12000359 DOI: 10.1038/s41598-025-96823-9] [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: 01/03/2025] [Accepted: 04/01/2025] [Indexed: 04/17/2025] Open
Abstract
Preeclampsia results from placental insufficiency and causes maternal endothelial dysfunction and multi-organ damage. Our in-silico analysis identified Cystatin 6 (CST6), a cysteine protease inhibitor, as located on the placental surface where it might be released into maternal circulation. This study aimed to characterise CST6 and one of its high affinity targets, Legumain (LGMN), in preeclampsia and assess its biomarker potential by measuring levels in maternal circulation. Placental CST6 mRNA expression was significantly increased in 78 pregnancies complicated by early-onset preeclampsia (delivering at < 34 weeks' gestation) relative to 30 gestation matched controls (P < 0.0001). LGMN mRNA expression was significantly decreased (P = 0.0309). Circulating CST6 was increased in 35 pregnancies complicated by early-onset preeclampsia (< 34 weeks' gestation) relative to 27 gestation matched controls (P = 0.0261), and LGMN levels remained unchanged. At 36 weeks' gestation, circulating CST6 was significantly increased (P = 0.001), while LGMN was significantly decreased (P = 0.0135) in 21 pregnancies preceding diagnosis of preeclampsia at term, compared to 184 pregnancies that did not develop preeclampsia. Human trophoblast stem cells (hTSC) were differentiated into syncytiotrophoblast or extravillous trophoblast (EVT) to evaluate CST6 and LGMN expression in these trophoblast lineages. CST6 and LGMN mRNA expression were significantly increased across 96 h after syncytiotrophoblast (P = 0.0066 and P = 0.0010 respectively) and EVT differentiation (P = 0.0618 and P = 0.0016 respectively), with the highest expression in syncytiotrophoblast. Computational analysis of two publicly available single-cell and single-nuclei RNA sequencing datasets correlated with the expression pattern observed in vitro. When syncytiotrophoblast cells were exposed to hypoxia (1% O2 vs. 8% O2), CST6 expression significantly increased (P = 0.0079), whilst LGMN expression was unchanged. The vascular endothelium may serve as an additional source of circulating CST6 and LGMN in preeclampsia. Induction of dysfunction in endothelial cells by TNFα, caused reduced CST6 expression (P = 0.0036), whilst LGMN expression remained unchanged. Administering recombinant CST6 to endothelial cells enhanced markers of endothelial dysfunction and LGMN expression in the presence of TNFα. These findings indicate an inverse relationship between CST6 and LGMN in the placenta and maternal circulation in preeclampsia. We suggest elevated circulating levels of CST6 may be induced by placental hypoxia. This study provides novel insight into the dysregulation of CST6 and LGMN in preeclampsia and introduces their potential roles in human pregnancy and associated pathology.
Collapse
Affiliation(s)
- Stefan M Botha
- Translational Obstetrics Group, The Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, 3084, Australia.
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia.
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Experimental and Clinical Research Center, a cooperation between Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany.
| | - Lucy A Bartho
- Translational Obstetrics Group, The Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Sunhild Hartmann
- Translational Obstetrics Group, The Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Ping Cannon
- Translational Obstetrics Group, The Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Anna Nguyen
- Translational Obstetrics Group, The Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Tuong-Vi Nguyen
- Translational Obstetrics Group, The Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Natasha Pritchard
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Ralf Dechend
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Department of Cardiology and Nephrology, HELIOS Clinic, Berlin, Germany
| | - Olivia Nonn
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Stephen Tong
- Translational Obstetrics Group, The Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Translational Obstetrics Group, The Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| |
Collapse
|
3
|
Toda S, Ikeda M, Suzuki T. Clinical utility of sFlt-1 and PlGF for transient hypertension and proteinuria in late pregnancy. CEN Case Rep 2025:10.1007/s13730-025-00994-3. [PMID: 40220070 DOI: 10.1007/s13730-025-00994-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Affiliation(s)
- Sayaka Toda
- Department of Nephrology, Kameda Medical Center, 296-8602, 929, Higashicho, Kamogawa, Chiba, Japan
- Department of Family Medicine, Kameda Family Clinic Tateyama, Chiba, Japan
| | - Mari Ikeda
- Department of Nephrology, Kameda Medical Center, 296-8602, 929, Higashicho, Kamogawa, Chiba, Japan
| | - Tomo Suzuki
- Department of Nephrology, Kameda Medical Center, 296-8602, 929, Higashicho, Kamogawa, Chiba, Japan.
| |
Collapse
|
4
|
Ling Y, Wu H, Li J, Ma Y, Zhang S, Lu Y, Feng J, Wang S, Liang X. Predictive value of maternal serum placental growth factor, maternal clinical features, and fetal ultrasound indicators in preeclampsia. BMC Pregnancy Childbirth 2025; 25:390. [PMID: 40181312 PMCID: PMC11969946 DOI: 10.1186/s12884-025-07517-z] [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: 02/06/2025] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
OBJECTIVE This study was aimed to evaluate the predictive value of placental growth factor (PLGF), maternal risk factors, and maternal-fetal ultrasound indicators for preeclampsia(PE) and to analyze delivery outcomes associated with PE. METHODS Pregnant women with PE(n = 80) and healthy controls(n = 160) were enrolled from the People's Hospital of Guangxi Zhuang Autonomous Region between 2021 and 2022. Serum PLGF level were measured at various stages of pregnancy: early pregnancy, mid-pregnancy, and late pregnancy. Maternal clinical features, fetal ultrasound indicators, and pregnancy outcomes were also recorded. RESULTS The serum concentration of PLGF was lower in the PE group compared to the control group throughout early, middle, and late pregnancy. At 20-24 weeks, the uterine artery resistance index was higher in the preeclampsia group (P < 0.05). Logistic regression analysis identified PLGF concentrations in early pregnancy and mid-pregnancy, along with mean arterial pressure (MAP), as independent factors for the onset of PE (P < 0.05). CONCLUSION PLGF could be used for prediction of PE in early pregnancy. A predictive model for PE was established by combining the PLGF concentrations in early and middle pregnancy with MAP.
Collapse
Affiliation(s)
- Yuee Ling
- Obstetrics Department, The People's Hospital of Guangxi Zhuang Autonomous Region, No.6 Taoyuan Road, Qingxiu District, Nanning City, Guangxi Province, China
| | - Hua Wu
- Obstetrics Department, The People's Hospital of Guangxi Zhuang Autonomous Region, No.6 Taoyuan Road, Qingxiu District, Nanning City, Guangxi Province, China
| | - Jing Li
- Obstetrics Department, The People's Hospital of Guangxi Zhuang Autonomous Region, No.6 Taoyuan Road, Qingxiu District, Nanning City, Guangxi Province, China
| | - Yanhua Ma
- Obstetrics Department, The People's Hospital of Guangxi Zhuang Autonomous Region, No.6 Taoyuan Road, Qingxiu District, Nanning City, Guangxi Province, China
| | - Shanshan Zhang
- Obstetrics Department, The People's Hospital of Guangxi Zhuang Autonomous Region, No.6 Taoyuan Road, Qingxiu District, Nanning City, Guangxi Province, China
| | - Yanqun Lu
- Obstetrics Department, The People's Hospital of Guangxi Zhuang Autonomous Region, No.6 Taoyuan Road, Qingxiu District, Nanning City, Guangxi Province, China
| | - Juan Feng
- Obstetrics Department, The People's Hospital of Guangxi Zhuang Autonomous Region, No.6 Taoyuan Road, Qingxiu District, Nanning City, Guangxi Province, China
| | - Sumei Wang
- Obstetrics Department, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning City, Guangxi Province, China.
| | - Xuxia Liang
- Obstetrics Department, The People's Hospital of Guangxi Zhuang Autonomous Region, No.6 Taoyuan Road, Qingxiu District, Nanning City, Guangxi Province, China.
| |
Collapse
|
5
|
de Jesús NR, de Jesús GR, Lacerda MI, Dos Santos FC, Nogueira JDS, Porto LC, Coutinho ESF, Klumb EM. Analysis of the Longitudinal Behavior of Serum Levels of Soluble Flt1 and Placental Growth Factor in Pregnant Patients With Systemic Lupus Erythematosus. Arthritis Care Res (Hoboken) 2025. [PMID: 40176387 DOI: 10.1002/acr.25536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 03/07/2025] [Accepted: 03/20/2025] [Indexed: 04/04/2025]
Abstract
OBJECTIVE This study analyzed longitudinal trajectories of soluble Flt1 (sFlt1) levels, placenta growth factor (PlGF) levels, and sFlt1:PlGF ratios in a cohort of pregnant patients with systemic lupus erythematosus (SLE). METHODS Blood samples were collected (14-18, 24-26, 30-32, 34-36, and 38-40 weeks), stored at -80°C, and evaluated for serum levels of sFlt1, PlGF, and sFlt1:PlGF ratios. Patients were classified as inactive SLE (Systemic Lupus Erythematosus Pregnancy Disease Activity Index [SLEPDAI] <4), active disease (SLEPDAI ≥4), or preeclampsia (PE). Medians and interquartile ranges were calculated for each group, and linear models with random effects were used. RESULTS A total of 527 samples were obtained from 163 patients, and all patients were subsequently classified as having inactive disease (109 patients [66.9%]), active disease (33 patients [20.2%]), and inactive disease with PE (21 patients [12.9%]). In exploratory analysis, patients with PE had higher mean serum levels of sFlt1 and sFlt1:PlGF ratios and lower PlGF levels than patients with inactive and active SLE (P = 0.01 to P < 0.001). Using linear models with random effects, there was no significant differences in mean serum levels of these angiogenic markers comparing inactive and active disease. Patients with PE showed a marked increase in sFlt1 levels from the 24th week, constantly low PlGF levels from the 14th week, and progressive increase of sFlt1:PlGF ratio during pregnancy. All these differences were statistically significant compared to the groups without PE. CONCLUSION Pregnant patients with SLE who developed PE had higher sFlt1 levels and sFlt1:PlGF ratios and lower PlGF levels, and these last two changes were detected at the beginning of second trimester, before clinical manifestation. SLE activity did not interfere with longitudinal behavior of these angiogenic markers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Evandro M Klumb
- Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
6
|
Cavoretto PI, Nayak NR, Odibo AO. Time to reconcile the dichotomy of the cardiovascular-placental axis. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2025; 65:401-403. [PMID: 40168639 DOI: 10.1002/uog.29207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/15/2025] [Indexed: 04/03/2025]
Abstract
Linked article: This Editorial comments on the article by Nan et al. Click here to view the article.
Collapse
Affiliation(s)
- P I Cavoretto
- Department of Obstetrics and Gynaecology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - N R Nayak
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, University of Missouri-Kansas City, Kansas City, MO, USA
| | - A O Odibo
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, University of Missouri-Kansas City, Kansas City, MO, USA
| |
Collapse
|
7
|
Burns LP, Potchileev S, Mueller A, Azzi M, Premkumar A, Peterson J, Rausch A, Gonzalez M, Silasi M, Karumanchi SA, Thadhani R, Rana S. Real-world evidence for the utility of serum soluble fms-like tyrosine kinase 1/placental growth factor test for routine clinical evaluation of hospitalized women with hypertensive disorders of pregnancy. Am J Obstet Gynecol 2025; 232:385.e1-385.e21. [PMID: 39029547 DOI: 10.1016/j.ajog.2024.07.015] [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: 05/22/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND An imbalance of the antiangiogenic factor, soluble fms-like tyrosine kinase-1, and proangiogenic factor, placental growth factor, in the circulation is a reliable predictor for the development of preeclampsia with severe features and related adverse outcomes. In 2023, the US Food and Drug Administration approved a serum soluble fms-like tyrosine kinase-1/placental growth factor test at a cutoff of 40 to aid in the risk assessment of women hospitalized for hypertensive disorders of pregnancy for the progression to preeclampsia with severe features between 23 and 35 weeks. OBJECTIVE This study aimed to generate real-world evidence for clinical utility for serum soluble fms-like tyrosine kinase-1/placental growth factor test when made available to clinicians in a timely fashion as an aid in risk stratification of development of preeclampsia with severe features within 2 weeks of testing among hospitalized patients with hypertensive disorders of pregnancy. STUDY DESIGN Hospitalized patients with hypertensive disorders of pregnancy between 23 weeks to 34 weeks and 6 days of gestation were prospectively studied from June 2023 to January 2024 after the implementation of serum soluble fms-like tyrosine kinase-1/placental growth factor testing into routine clinical practice. Serum samples were obtained from patients via venipuncture and analyzed on an automated immunoassay platform (placental growth factor and soluble fms-like tyrosine kinase-1 assays; Thermo Fisher Scientific). Before implementation, physicians were educated on appropriate use and management guidelines on the basis of biomarkers but made pragmatic management decisions independently. Results of soluble fms-like tyrosine kinase-1/placental growth factor tests were available to clinicians within 24 hours of venipuncture. The association between soluble fms-like tyrosine kinase-1/placental growth factor ≥40 and progression to preeclampsia with severe features and adverse maternal/perinatal outcomes were assessed. RESULTS Of the 65 patient encounters, 36 had a soluble fms-like tyrosine kinase-1/placental growth factor <40 (55.4%). The rate of delivery for indications related to hypertensive disorders of pregnancy within 2 weeks was significantly lower among encounters with a low ratio vs high ratio (2/36 [5.6%] vs 21/29 [72.4%]) even after controlling for relevant confounders (adjusted hazard ratio, 7.52; 95% confidence interval, 3.05-18.54; P<.001). A diagnosis of preeclampsia with severe features within 2 weeks of testing was also less likely among the encounters with soluble fms-like tyrosine kinase-1/placental growth factor ratio <40 when compared with soluble fms-like tyrosine kinase-1/placental growth factor ratio ≥40 (2/36 [5.6%] vs 23/29 [79.3%], P<.001; positive predictive value of 79% [95% confidence interval, 0.65-0.94] and negative predictive value of 0.94 [95% confidence interval, 0.87-1.00]). The positive and negative likelihood ratios for the development of preeclampsia with severe features within 2 weeks of testing were 6.13 and 0.09, respectively. Encounters with a soluble fms-like tyrosine kinase-1/placental growth factor ratio <40 were less likely to experience a maternal or fetal adverse event as compared with encounters with soluble fms-like tyrosine kinase-1/placental growth factor ratio ≥40 (3/36 [8.3%] vs 10/29 [34.5%], P=.01). Among 36 encounters involving low soluble fms-like tyrosine kinase-1/placental growth factor values, 22 had had equivocal clinical or laboratory criteria resembling preeclampsia at presentation but were expectantly managed on the basis of biomarkers, and none developed preeclampsia with severe features or adverse outcomes at 2 weeks. The median latency defined as days between biomarker measurement and delivery in patients with a low biomarker ratio was 33 (interquartile ratio, 23-47) vs 7 (interquartile ratio, 4-14) days among patients with a high ratio (P<.001). Corticosteroid use within 2 weeks was also significantly reduced in the low biomarker group when compared with the high biomarker group (8/35 [22.9%] vs 24/29 [82.8%], P<.001). CONCLUSION In this study, the incorporation of soluble fms-like tyrosine kinase-1/placental growth factor ratio into clinical practice serves as a dependable supplement in assessing risk for progression to preeclampsia with severe features and adverse outcomes in patients with hypertensive disorders of pregnancy in the United States. Among patients with a low ratio, pregnancy may be prolonged, which results in better neonatal outcomes without harm to the mother.
Collapse
Affiliation(s)
- Luke P Burns
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago Medical Center, Chicago, IL
| | - Sanela Potchileev
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago Medical Center, Chicago, IL
| | - Ariel Mueller
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago Medical Center, Chicago, IL; Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA
| | - Marly Azzi
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago Medical Center, Chicago, IL
| | - Ashish Premkumar
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago Medical Center, Chicago, IL
| | - Jessica Peterson
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago Medical Center, Chicago, IL
| | - Andrew Rausch
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago Medical Center, Chicago, IL
| | - Maritza Gonzalez
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago Medical Center, Chicago, IL
| | - Michelle Silasi
- Division of Maternal Fetal Medicine, Department of Obstetrics & Gynecology, Mercy Hospital, St. Louis, MO
| | | | | | - Sarosh Rana
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago Medical Center, Chicago, IL.
| |
Collapse
|
8
|
Businge CB, Longo-Mbenza B, Kengne AP. Circulating Potassium/Magnesium Ratio, Thyroid Stimulating Hormone, Fasting Plasma Glucose, Oxidized LDL/Albumin Ratio, and Urinary Iodine Concentration Are Possible Entities for Screening for Preeclampsia in Low-Resource Settings. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:600. [PMID: 40282890 PMCID: PMC12028386 DOI: 10.3390/medicina61040600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/20/2025] [Accepted: 03/23/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: Several micro- and macro-nutrient malnutrition states that are routinely assessed during clinical care of women in the antenatal period have been proposed as risk factors for preeclampsia. However, there is a paucity of data on the potential use of these biomarkers for detection of preeclampsia. The aim of this case-control study was to investigate the association of biomarkers from routine clinical tests, and those specific to micro- and macro-nutrient malnutrition, with the risk of preeclampsia. Materials and Methods: Venous blood samples of 250 participants with preeclampsia and 150 pregnant women without preeclampsia were collected and assayed immediately for the full blood count, urea and electrolytes, high-density cholesterol (HDL), total cholesterol, triglycerides, low-density lipoprotein cholesterol (LDL), oxidized low-density lipoprotein cholesterol (OxLDL), and selenium, in addition to urine iodine concentration (UIC). Results: The serum potassium/magnesium ratio (K+/Mg2+), UIC, fasting plasma glucose (FPG), thyroid stimulating hormone (TSH), lymphocyte percentage (L/WBC%), and the oxidized LDL/albumin ratio (OxLDL/Alb) were identified as independent predictors of preeclampsia. Conclusions: Serum potassium/magnesium ratio and other analytes essential for various biological processes, some of which are assayed during routine care, were significantly associated with preeclampsia, warranting further exploration as potential screening biomarkers in low-resource settings.
Collapse
Affiliation(s)
- Charles Bitamazire Businge
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health Sciences, Walter Sisulu University, Mthatha 5117, South Africa
| | - Benjamin Longo-Mbenza
- Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of the Congo;
- Department of Public Health, Lomo University of Research, 652 Freesias, Kinshasa, Democratic Republic of the Congo
| | | |
Collapse
|
9
|
Tian S, He L, Pan A, Zhang L, Wang J. sFlt-1/PlGF ratio combined with endocan-1 serum levels improves the predictive values for the occurrence and prognosis of preeclampsia in a single centre study. J Hum Hypertens 2025:10.1038/s41371-025-01009-5. [PMID: 40133446 DOI: 10.1038/s41371-025-01009-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/18/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025]
Abstract
This retrospective study evaluated diagnostic and prognostic values of sFlt-1/PlGF ratio combined with serum endocan-1 for preeclampsia (PE). This study included 105 patients with PE admitted at Xiamen University Affiliated Xiang'an Hospital from January 2020 to September 2023, with 90 healthy pregnant women receiving routine check-ups during the same period as controls. sFlt-1, PlGF, and endocan-1 levels were measured, and sFlt-1/PlGF ratio was calculated. The correlation of sFlt-1/PlGF ratio with serum endocan-1 levels was analysed, and influencing factors for poor prognosis of PE patients were screened. Diagnostic and prognostic values of sFlt-1/PlGF ratio combined with serum endocan-1 were assessed. Results showed an increase in sFlt-1/PlGF ratio and serum endocan-1 levels in PE patients and a positive correlation between them. For assisting in the diagnosis of PE, the AUC of the sFlt-1/PlGF ratio combined with serum endocan-1 was 0.874 (95%CI: 0.819 - 0.917; sensitivity, 75.24%; specificity, 100.00%), which was superior over that of sFlt-1/PlGF ratio (P = 0.025) and endocan-1 (P = 0.047) alone. Elevated sFlt-1/PlGF ratio and serum endocan-1 levels were independent risk factors for poor prognosis of PE patients. For assisting in predicting poor prognosis of PE, the AUC of sFlt-1/PlGF ratio with serum endocan-1 was 0.955 (95%CI: 0.896 - 0.986; sensitivity, 82.61%; specificity, 100.00%), which was higher than that of sFlt-1/PlGF ratio (P = 0.023) or serum endocan-1 (P = 0.010) alone. Altogether, sFlt-1/PlGF ratio combined with serum endocan-1 is advantageous over sFlt-1/PlGF ratio and serum endocan-1 alone for predicting PE occurrence and prognosis.
Collapse
Affiliation(s)
- Shanshan Tian
- Department of Obstetrics and Gynecology, Xiamen University Affiliated Xiang'an Hospital, No.2000 Xiang'an East Road, Xiang'an District, Xiamen City, 361000, China
| | - Libin He
- Chiropractic Room, Xiamen University Affiliated Xiang'an Hospital, No.2000 Xiang'an East Road, Xiang'an District, Xiamen City, 361000, China
| | - Ahuang Pan
- Department of Obstetrics and Gynecology, Xiamen University Affiliated Xiang'an Hospital, No.2000 Xiang'an East Road, Xiang'an District, Xiamen City, 361000, China
| | - Lina Zhang
- Department of Obstetrics and Gynecology, Xiamen University Affiliated Xiang'an Hospital, No.2000 Xiang'an East Road, Xiang'an District, Xiamen City, 361000, China
| | - Jielin Wang
- Department of Obstetrics and Gynecology, Xiamen University Affiliated Xiang'an Hospital, No.2000 Xiang'an East Road, Xiang'an District, Xiamen City, 361000, China.
| |
Collapse
|
10
|
Tonni G, Lituania M, Grisolia G, Pinto A, Bonasoni MP, Rizzo G, Ruano R, Araujo Júnior E, Werner H, Sepulveda W, Pilu G. Placental and Umbilical Cord Anomalies Detected by Ultrasound as Clinical Risk Factors of Adverse Perinatal Outcomes. Case Series Review of Selected Conditions. Part 3: Vascular Anomalies of the Umbilical Cord and Fetoplacental Vascular Malperfusion. JOURNAL OF CLINICAL ULTRASOUND : JCU 2025. [PMID: 40035182 DOI: 10.1002/jcu.23904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 03/05/2025]
Abstract
In the previous published reviews Part 1 and Part 2, we examined the association between placental and umbilical cord anomalies in relation to adverse perinatal outcomes. In this conclusive Part 3, only umbilical cord vascular anomalies are considered, together with the perinatal effects caused by maternal vascular malperfusion and the secondary fetal vascular malperfusion anomalies. Specifically, the review comprises the following umbilical cord pathologies: umbilical cord torsion/stricture, amniotic band syndrome and umbilical cord strictures/strangulation, umbilical cord hemorrhagic cyst, umbilical cord rupture/cord hemangioma, umbilical cord hematoma, and umbilical cord ulceration. A series of case presentations and a gallery of images have been included to illustrate this final review.
Collapse
Affiliation(s)
- Gabriele Tonni
- Department of Obstetrics and Neonatology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), AUSL Reggio Emilia, Reggio Emilia, Italy
| | - Mario Lituania
- Preconceptional and Prenatal Pathophysiology, Department of Obstetrics and Gynecology, E.O. Ospedali Galliera, Genoa, Italy
| | - Gianpaolo Grisolia
- Department of Obstetrics and Gynecology, Carlo Poma Hospital, AST Mantova, Mantua, Italy
| | - Alessia Pinto
- Department of Obstetrics and Gynecology, Carlo Poma Hospital, AST Mantova, Mantua, Italy
| | - Maria Paola Bonasoni
- Department of Pathology, Santa Maria Nuova Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), AUSL Reggio Emilia, Reggio Emilia, Italy
| | - Giuseppe Rizzo
- Department of Obstetrics and Urologic Sciences, Policlinic Hospital Umberto I, University La Sapienza, Rome, Italy
| | - Rodrigo Ruano
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Edward Araujo Júnior
- Department of Obstetrics, Paulista School of Medicine - Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Heron Werner
- Department of Fetal Medicine, Biodesign Laboratory DASA/PUC, Rio de Janeiro, Brazil
| | - Waldo Sepulveda
- FETALMED-Maternal-Fetal Diagnostic Center, Fetal Imaging Unit, Santiago, Chile
| | - Gianluigi Pilu
- Department of Obstetrics and Gynecology, Policlinic Hospital Sant'Orsola, University of Bologna, Bologna, Italy
| |
Collapse
|
11
|
Shen X, Tao Y, Wang Y, Obore N, Yu H. Research Hotspots and Thematic Trends in the Management of Preeclampsia: A Bibliometric Analysis from 2000 to 2022. Reprod Sci 2025; 32:815-824. [PMID: 39741168 DOI: 10.1007/s43032-024-01773-8] [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: 07/22/2023] [Accepted: 12/16/2024] [Indexed: 01/02/2025]
Abstract
Preeclampsia (PE) is one of the serious complications of pregnancy, and the management of PE remains an important problem for obstetricians. This study aims to identify the characteristics and trends of published articles on PE management through bibliometric analysis. We searched the Web of Science database for articles related to PE management from 2000 to 2022. Metadata was obtained, including the titles, publication dates, authors, institutions, countries, and keywords of all articles, and then network visualization and burst keyword analysis were performed using Citespace and VOSviewer software. A total of 5190 articles were included in the analysis. The number of publications in the field of PE management has steadily increased over the years, and a visual analysis of collaborative networks of authors, institutions, and countries revealed that the United States, United Kingdom, Australia, and Canada have contributed the most to the field and formed extensive collaborations. The Journal of maternal-fetal & neonatal medicine has the most publications in this field, and the Journal of Obstetrics and gynecology has not only more publications but also 64.75 citations per article. The keywords mainly focused on prevention, diagnosis, risk factors, and outcome of PE. In addition, hypertensive disorders of pregnancy and fetal growth restriction have received a lot of attention in this field in recent years. We analyzed the partnerships in the field of PE management through bibliometrics and showed trends and developments in the field. The available results suggest that PE management will continue to be a focus of attention for obstetricians and researchers.
Collapse
Affiliation(s)
- Xiao Shen
- Department of Obstetrics and Gynecology, Southeast University Affiliated Zhongda Hospital, Nanjing, Jiangsu, 210009, China
| | - Yuchen Tao
- Southeast University Medical College, Nanjing, China
| | - Yixiao Wang
- Southeast University Medical College, Nanjing, China
| | - Nathan Obore
- Southeast University Medical College, Nanjing, China
| | - Hong Yu
- Department of Obstetrics and Gynecology, Southeast University Affiliated Zhongda Hospital, Nanjing, Jiangsu, 210009, China.
| |
Collapse
|
12
|
Giardini V, Pelucchi A, Daolio C, Casati M, Vergani P, Lattuada M, Locatelli A. Coffin-Siris Syndrome and Unusual Angiogenic Profiles in Pregnancy: A Case Study Emphasizing Caution in Interpreting a Very Low sFlt-1/PlGF Ratio. Am J Med Genet A 2025; 197:e63939. [PMID: 39535382 DOI: 10.1002/ajmg.a.63939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/15/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Valentina Giardini
- Department of Obstetrics, IRCCS San Gerardo Dei Tintori Foundation, University of Milano-Bicocca, Monza, Italy
| | - Arianna Pelucchi
- Department of Obstetrics, IRCCS San Gerardo Dei Tintori Foundation, University of Milano-Bicocca, Monza, Italy
| | - Cecilia Daolio
- Pediatrics, IRCCS San Gerardo Dei Tintori Foundation, Monza, Italy
| | - Marco Casati
- Laboratory Medicine, IRCCS San Gerardo Dei Tintori Foundation, Monza, Italy
| | - Patrizia Vergani
- Department of Obstetrics, MBBM Foundation Onlus, University of Milano-Bicocca, Monza, Italy
| | - Martina Lattuada
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Anna Locatelli
- Department of Obstetrics, IRCCS San Gerardo Dei Tintori Foundation, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
13
|
Lundgaard MH, Sinding MM, Sørensen AN, Torp NMU, Handberg A, Andersen S, Andersen SL. Maternal Thyroid Function and Biochemical Markers of Placental Function in Early Pregnancy. Clin Endocrinol (Oxf) 2025; 102:306-314. [PMID: 39370704 DOI: 10.1111/cen.15145] [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: 07/01/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 10/08/2024]
Abstract
OBJECTIVE A link between maternal thyroid function and the placental biomarkers, soluble fms-like tyrosine kinase-1 (sFlt-1) and placental growth factor (PlGF), has been brought forward. This study aimed to describe their association in early pregnancy. DESIGN Retrospective cohort study. PARTICIPANTS Eight hundred and fifty-eight pregnant women from the North Denmark Region, 2013, with blood samples drawn in early pregnancy. MEASUREMENTS Thyroid-stimulating hormone (TSH), free thyroxine (fT4), thyroid-peroxidase antibodies (TPO-Ab), thyroglobulin antibodies (Tg-Ab) (ADVIA Centaur XPT, Siemens Healthineers), sFlt-1 and PlGF (Kryptor Compact, ThermoFisher Scientific) were measured. The association between maternal TSH and fT4 and percentile (pc) levels of sFlt-1 and PlGF (< 25th pc, 25-75th pc, > 75th pc) was evaluated using regression analysis and reported as adjusted beta coefficient (aβ). The frequency of maternal thyroid autoantibodies (TPO-Ab > 60 U/mL or Tg-Ab > 33 U/mL) by pc levels of sFlt-1 and PlGF was compared using chi-squared test. RESULTS Higher levels (> 75th pc) of sFlt-1 associated with lower TSH (aβ 0.62, 95% CI: 0.51-0.76) and higher fT4 (aβ 1.03, 95% CI: 1.01-1.05). Higher levels of PlGF associated with lower TSH (aβ 0.82, 95% CI: 0.69-0.98), but not with levels of fT4 (aβ 1.00, 95% CI: 0.97-1.02). No association with maternal thyroid autoantibodies was found (TPO-Ab: sFlt-1: p-value 0.5 and PlGF: p-value 0.1; Tg-Ab: sFlt-1: p-value 0.7 and PlGF: p-value 0.1). CONCLUSIONS In a large cohort of Danish pregnant women, higher levels of sFlt-1 and PlGF associated with maternal thyroid function in early pregnancy, while there was no association with maternal thyroid autoantibodies.
Collapse
Affiliation(s)
- Maja H Lundgaard
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Marianne M Sinding
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Obstetrics and Gynecology, Aalborg University Hospital, Aalborg, Denmark
| | - Anne N Sørensen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Obstetrics and Gynecology, Aalborg University Hospital, Aalborg, Denmark
| | - Nanna M U Torp
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Stig Andersen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Geriatrics, Aalborg University Hospital, Aalborg, Denmark
| | - Stine L Andersen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
14
|
Shajihan A, Honigberg MC, Saad A, Scott NS, Powell-Wiley TM, Sharma G. Is it Time for Precision Screening in Preeclampsia? JACC. ADVANCES 2025; 4:101585. [PMID: 39938183 PMCID: PMC11870246 DOI: 10.1016/j.jacadv.2025.101585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/13/2024] [Indexed: 02/14/2025]
Affiliation(s)
- Ain Shajihan
- Inova Schar Heart and Vascular Institute, Inova Health System, Falls Church, Virginia, USA.
| | - Michael C Honigberg
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Antonio Saad
- Department of Maternal Fetal Medicine, Inova Health System, Inova Women's and Children's Hospital, Falls Church, Virginia, USA
| | - Nandita S Scott
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Tiffany M Powell-Wiley
- Cardiovascular Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA; Intramural Research Program, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, Maryland, USA
| | - Garima Sharma
- Inova Schar Heart and Vascular Institute, Inova Health System, Falls Church, Virginia, USA
| |
Collapse
|
15
|
Liao L, Liu M, Yin Y, Xu Q, Yuan L, Xie S, Zhou R. Glycosylated fibronectin in preeclampsia: a systematic review and meta-analysis. BMC Pregnancy Childbirth 2025; 25:228. [PMID: 40022002 PMCID: PMC11871604 DOI: 10.1186/s12884-025-07243-6] [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: 03/26/2024] [Accepted: 01/28/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Preeclampsia (PE) is a complex multisystem disease, and its timely diagnosis and treatment impact the health of patients and perinatal infants. Studies have reported elevated levels of maternal serum glycosylated fibronectin (GlyFn) in patients with PE compared with pregnant women without PE (controls), suggesting its potential as a novel biomarker for screening and diagnosing PE. Therefore, this study aims to evaluate maternal serum GlyFn levels and their diagnostic accuracy in PE. METHODS A systematic literature search was conducted using PubMed, EMBASE, Web of Science, and the Cochrane Library up to January 15, 2024. The Newcastle-Ottawa Quality Assessment Scale and the Quality Assessment of Diagnostic Accuracy Studies-2 tool were used to evaluate study quality. Heterogeneity was assessed using I2 statistics. In the meta-analysis comparing maternal serum GlyFn levels between PE and controls, standardized mean differences (SMDs) and 95% confidence intervals (CIs) were calculated. Publication bias was detected, and sensitivity and subgroup analyses were conducted to verify the robustness of the results and identify potential sources of heterogeneity. For the meta-analysis of diagnostic, the accuracy of maternal serum GlyFn levels between PE and controls, sensitivity and specificity were pooled and the summary receiver operating characteristic curve and area under the curve were used as measures of overall accuracy. This review was registered in the International Prospective Register of Systematic Reviews (registration number: CRD42024512172). RESULTS A total of 11 studies were included, and the meta-analysis revealed that maternal serum GlyFn levels were significantly higher in the PE group than in the control group (SMD = 1.08, 95% CI = 0.72-1.43, P < 0.001). Heterogeneity may arise from differences in the detection method and research type. Overall, the combined sensitivity and specificity of maternal serum GlyFn levels in diagnosing PE were 0.81 (95% CI = 0.77-0.85, P < 0.001) and 0.80 (95% CI = 0.77-0.82, P < 0.001), respectively, with an area under the curve of 0.90. CONCLUSIONS This meta-analysis confirmed that maternal serum GlyFn levels are significantly higher in patients with PE and exhibit high diagnostic accuracy for PE diagnosis, suggesting its potential as a biomarker for screening and diagnosing PE.
Collapse
Affiliation(s)
- Lingyun Liao
- Department of Obstetrics and Gynecology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, (Sichuan University) of Ministry of Education, Chengdu, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Min Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, (Sichuan University) of Ministry of Education, Chengdu, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Yangxue Yin
- Department of Obstetrics and Gynecology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, (Sichuan University) of Ministry of Education, Chengdu, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Qin Xu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, (Sichuan University) of Ministry of Education, Chengdu, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Liming Yuan
- Department of Obstetrics and Gynecology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, (Sichuan University) of Ministry of Education, Chengdu, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Shuangshuang Xie
- Department of Obstetrics and Gynecology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, (Sichuan University) of Ministry of Education, Chengdu, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Rong Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, (Sichuan University) of Ministry of Education, Chengdu, China.
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China.
| |
Collapse
|
16
|
Zhao Y, Xu L, An P, Zhou J, Zhu J, Liu S, Zhou Q, Li X, Xiong Y. A nomogram for predicting adverse perinatal outcome with fetal growth restriction: a prospective observational study. BMC Pregnancy Childbirth 2025; 25:132. [PMID: 39934709 DOI: 10.1186/s12884-025-07252-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 01/28/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Fetal growth restriction (FGR) is a major determinant of perinatal morbidity and mortality. Our study aimed to develop a prediction model for the risk of FGR developing adverse perinatal outcome (APO) and evaluate its performance. METHODS This was a prospective observational cohort study of consecutive singleton gestations meeting the ACOG-endorsed criteria for FGR from January 2022 to June 2023 at Obstetrics and Gynecology Hospital of Fudan University. Clinical information, ultrasound indicators and serum biomarkers were collected. The primary composite APO comprised one or more of: perinatal death, intrauterine demise, intraventricular hemorrhage, periventricular leukomalacia, seizures, necrotizing enterocolitis, neonatal respiratory distress syndrome, sepsis and the length of stay in the neonatal intensive care unit > 7 days. Least absolute shrinkage and selection operator regression was used to screen variables for nomogram model construction. The discrimination, calibration and clinical effectiveness of the nomogram were evaluated using receiver operating characteristic curve, calibration plots and decision curve analysis in training and validation cohorts. RESULTS A total of 122 pregnancies were enrolled in the final statistical analysis. Five variables were identified to establish a nomogram, including gestational weeks at diagnosis, abnormal umbilical artery Doppler, abnormal uterine artery Doppler, and multiples of the median values of placental growth factor and soluble fms-like tyrosine kinase-1. The area under the receiver-operating-characteristics curve of 0.87 (95% CI, 0.75-0.99) and 0.86 (95% CI, 0.74-0.98) in the training and validation cohort respectively, indicated satisfactory discriminative ability of the nomogram. The calibration plots showed favorable consistency between the nomogram's predictions and actual observations. Decision curve analysis supported its practical value in a clinical setting. CONCLUSIONS A nomogram was developed and validated to possess the promising capacity of predicting APO in FGR-afflicted neonates, and may prove useful in counseling and management of pregnancies complicated by FGR.
Collapse
Affiliation(s)
- Ying Zhao
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Lei Xu
- Chang Ning Maternity & Infant Health Hospital, Shanghai, China
| | - Ping An
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jizi Zhou
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jie Zhu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Shuangping Liu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Qiongjie Zhou
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xiaotian Li
- Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China.
| | - Yu Xiong
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
17
|
Ndarugendamwo AM, Schlatter B, Surbek D, Radan AP. Massively elevated sFlt1/PlGF in a pregnant patient with fetal growth restriction and perinatal death in the context of a systemic lupus erythematosus. BMJ Case Rep 2025; 18:e258131. [PMID: 39914866 PMCID: PMC11800068 DOI: 10.1136/bcr-2023-258131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/10/2025] [Indexed: 02/09/2025] Open
Abstract
We report a case of confusingly high soluble fms-like tyrosine kinase 1/placental growth factor (sFlt1/PlGF) ratio in a pregnant patient with underlying systemic lupus erythematosus and antiphospholipid syndrome without superimposed pre-eclampsia.
Collapse
Affiliation(s)
| | - Bettina Schlatter
- Department of Obstetrics and Gynecology, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Daniel Surbek
- Department of Obstetrics and Gynecology, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Anda-Petronela Radan
- Department of Obstetrics and Gynecology, University Hospital Bern, University of Bern, Bern, Switzerland
| |
Collapse
|
18
|
Nie Q, He F. Testing for routine clinical evaluation of hospitalized women with hypertensive disorders of pregnancy. Am J Obstet Gynecol 2025; 232:e70. [PMID: 39278339 DOI: 10.1016/j.ajog.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Affiliation(s)
- Qingwen Nie
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510515, China
| | - Fang He
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510515, China.
| |
Collapse
|
19
|
Özkan S, Aksan A, Fıratlıgil FB, Kurt D, Sucu S, Coşkun A, Yücel KY, Çağlar AT, Üstün YE. Profilin-1 levels in preeclampsia: Associations with disease and adverse neonatal outcomes. Placenta 2025; 159:140-145. [PMID: 39721377 DOI: 10.1016/j.placenta.2024.12.012] [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: 09/11/2024] [Revised: 12/14/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Preeclampsia is a serious pregnancy complication requiring early detection to improve outcomes. Profilin-1 (PFN1), linked to vascular dysfunction, may serve as a biomarker for diagnosing preeclampsia and predicting adverse neonatal outcomes. The aim of this study was to determine the serum Profilin-1 levels in patients diagnosed with preeclampsia and to investigate its association with disease severity and adverse neonatal outcomes. METHODS A prospective cross-sectional study was conducted at Etlik City Hospital involving 40 women with preeclampsia and 40 healthy controls. Serum PFN1 levels were measured by ELISA and results were compared between groups. The results were compared between the groups. Receiver operating characteristic (ROC) curves were used to evaluate the diagnostic performance of PFN1. RESULTS Serum PFN1 levels were significantly higher in the preeclampsia group compared to controls (46.48 [30.23-60.29] vs. 26.41 [19.65-41.76], p < 0.001). The ROC curve showed good diagnostic accuracy for PFN1 in detecting preeclampsia with an AUC of 0.741 (95 % CI: 0.631-0.832, p < 0.001), a sensitivity of 95 % and a specificity of 42.5 %. PFN1 levels were also associated with composite neonatal outcomes, with an AUC of 0.622 (95 % CI: 0.520-0.716, p = 0.042). DISCUSSION PFN1 is a potential biomarker for the diagnosis of preeclampsia. However, further studies are needed to validate its role in predicting adverse neonatal outcomes and to improve its specificity for clinical use.
Collapse
Affiliation(s)
- Sadullah Özkan
- Division of Perinatology, Department of Obstetrics and Gynecology, Ankara Etlik City Hospital, Ankara, Turkey.
| | - Alperen Aksan
- Department of Obstetrics and Gynecology, Ankara Etlik Zubeyde Hanim Women's Health Education and Research Hospital, Ankara, Turkey
| | - Fahri B Fıratlıgil
- Division of Perinatology, Department of Obstetrics and Gynecology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Dilara Kurt
- Department of Obstetrics and Gynecology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Serap Sucu
- Department of Obstetrics and Gynecology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Aslıhan Coşkun
- Department of Obstetrics and Gynecology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Kadriye Yakut Yücel
- Division of Perinatology, Department of Obstetrics and Gynecology, Ankara Etlik City Hospital, Ankara, Turkey
| | - A Turhan Çağlar
- Division of Perinatology, Department of Obstetrics and Gynecology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Yaprak Engin Üstün
- Department of Obstetrics and Gynecology, Ankara Etlik Zubeyde Hanim Women's Health Education and Research Hospital, Ankara, Turkey
| |
Collapse
|
20
|
Jørgensen MM, Bæk R, Sloth JK, Sammour R, Sharabi-Nov A, Vatish M, Meiri H, Sammar M. A novel multiple marker microarray analyzer and methodology to predict major obstetric syndromes using surface markers of circulating extracellular vesicles from maternal plasma. Acta Obstet Gynecol Scand 2025; 104:151-163. [PMID: 39607297 DOI: 10.1111/aogs.15020] [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: 06/28/2024] [Revised: 10/21/2024] [Accepted: 11/10/2024] [Indexed: 11/29/2024]
Abstract
INTRODUCTION Placental-derived extracellular vesicles (EVs) are nano-organelles that facilitate intercellular communication between the feto-placental unit and the mother. We evaluated a novel Multiple Microarray analyzer for identifying surface markers on plasma EVs that predict preterm delivery and preeclampsia compared to term delivery controls. MATERIAL AND METHODS In this prospective exploratory cohort study pregnant women between 24 and 40 gestational weeks with preterm delivery (n = 16), preeclampsia (n = 19), and matched term delivery controls (n = 15) were recruited from Bnai Zion Medical Center, Haifa, Israel. Plasma samples were tested using a multiple microarray analyzer. Glass slides with 17 antibodies against EV surface receptors - were incubated with raw plasma samples, detected by biotinylated secondary antibodies specific to EVs or placental EVs (PEVs), and labeled with cyanine 5-streptavidin. PBS and whole human IgG served as controls. The fluorescent signal ratio to negative controls was log 2 transformed and analyzed for sensitivity and specificity using the area under the receiver operating characteristics curves (AUROC). Best pair ratios of general EVs/PEVs were used for univariate analysis, and top pairs were combined for multivariate analysis. Results were validated by comparison with EVs purified using standard procedures. RESULTS Heatmaps differentiated surface profiles of preeclampsia, preterm delivery, and term delivery receptors on total EVs and PEVs. Similar results were obtained with enriched EVs and EVs from raw plasma. Univariate analyses identified markers predicting preterm delivery and preeclampsia over term delivery controls with AUC >0.6 and sensitivity >50% at 80% specificity. Combining the best markers in a multivariate model, preeclampsia prediction over term delivery had an AUC of 0.89 (95% CI: 0.72-1.0) with 90% sensitivity and 90% specificity, marked by inflammation (TNF RII), relaxation (placenta protein 13 (PP13)), and immune-modulation (LFA1) receptors. Preterm delivery prediction over term delivery had an AUC of 0.97 (0.94-1.0), 84% sensitivity, and 90% specificity, marked by cell adhesion (ICAM), immune suppression, and general EV markers (CD81, CD82, and Alix). Preeclampsia prediction over preterm delivery had an AUC of 0.91 (0.79-0.99) with 80% sensitivity and 90% specificity with markers for complement activation (C1q) and autoimmunity markers. CONCLUSIONS The new, robust EV Multi-Array analyzer and methodology offer a simple, fast diagnostic tool that reveals novel surface markers for major obstetric syndromes.
Collapse
Affiliation(s)
- Malene Møller Jørgensen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Rikke Bæk
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Jenni K Sloth
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Rami Sammour
- Department of Obstetrics and Gynecology, Maternal and Fetal Medicine Unit, Bnai-Zion University Medical Center, Haifa, Israel
| | - Adi Sharabi-Nov
- Department of Statistics, Tel Hai Academic College, Tel Hai and Ziv Medical Center, Safed, Israel
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | | | - Marei Sammar
- Prof. Ephraim Katzir Department of Biotechnology Engineering, Braude College of Engineering, St, Karmiel, Israel
| |
Collapse
|
21
|
Feenstra ME, Schoots MH, Bezemer R, van der Meeren LE, Nikkels PG, Goor HV, Hillebrands JL, Prins JR, Ganzevoort W, Gordijn SJ. Placental histopathology in early-onset fetal growth restriction with use of sildenafil, a secondary analysis of the Dutch STRIDER study. Placenta 2025; 159:39-46. [PMID: 39616670 DOI: 10.1016/j.placenta.2024.10.024] [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: 08/16/2024] [Revised: 10/15/2024] [Accepted: 10/26/2024] [Indexed: 01/15/2025]
Abstract
OBJECTIVES Placental pathology lesions are common in early-onset fetal growth restriction (eoFGR). Therapeutic interventions to improve eoFGR outcomes are needed. In the international STRIDER trials (Sildenafil Therapy In Dismal prognosis Early-onset intrauterine growth Restriction) sildenafil didn't improve perinatal outcomes of eoFGR. We aimed to study the underlying placental pathology in the Dutch STRIDER trial and the effects of sildenafil on placental histopathology by describing the associations with sildenafil treatment, placental-dysfunction serum biomarkers and markers of oxidative stress. METHODS The Dutch STRIDER trial was a randomized controlled trial of sildenafil versus placebo in 216 singleton pregnancies complicated by eoFGR, included between 20+0- and 29+6-weeks' gestation. In 158 cases, placental histology was available. Lesions were classified independently by three perinatal pathologists blinded for clinical data, according to the international criteria of the Amsterdam Placental Workshop Group Consensus Statement. Blood samples taken at inclusion were analyzed for free thiols (FT), placental growth factor (PlGF) and soluble FMS-like tyrosine kinase-1 (sFlt-1, n = 85). RESULTS The 'big four' placental lesions (maternal and fetal vascular malperfusion, and chronic or acute inflammatory lesions) were equally distributed in both groups. However, massive perivillous fibrin deposition (MPFD) and chronic histiocytic intervillositis (CHIV) were less common in the sildenafil-treated group compared to the placebo-treated group (p = 0.026 and p = 0.043). FT, PlGF and sFlt-1 at inclusion were not discriminative for placental lesions. CONCLUSIONS Sildenafil had no effect on common placental lesions. The lower incidence of MPFD and CHIV after sildenafil exposure merits more research on the interaction between sildenafil and the immune system.
Collapse
Affiliation(s)
- Marjon E Feenstra
- Department of Obstetrics and Gynaecology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Mirthe H Schoots
- Department of Pathology & Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Romy Bezemer
- Department of Obstetrics and Gynaecology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Lotte-Elisabeth van der Meeren
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands; Department of Pathology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Peter Gj Nikkels
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Harry van Goor
- Department of Pathology & Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jan-Luuk Hillebrands
- Department of Pathology & Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jelmer R Prins
- Department of Obstetrics and Gynaecology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Wessel Ganzevoort
- Department of Obstetrics and Gynaecology, Amsterdam University Medical Center, Amsterdam, the Netherlands; Amsterdam Reproduction & Development Research Institute, Amsterdam, the Netherlands
| | - Sanne J Gordijn
- Department of Obstetrics and Gynaecology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
22
|
Zha Z, Jia C, Zhou R, Yin Q, Hu Y, Huang Z, Peng L, Zhang Y, Qiu X, Chen Y, Zhong Y, Wang Y, Pang M, Lu S, Sheng C, Huang L. Clostridium difficile-derived membrane vesicles promote fetal growth restriction via inhibiting trophoblast motility through PPARγ/RXRα/ANGPTL4 axis. NPJ Biofilms Microbiomes 2024; 10:158. [PMID: 39741137 DOI: 10.1038/s41522-024-00630-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/07/2024] [Indexed: 01/02/2025] Open
Abstract
Fetal growth restriction (FGR) is a common complication of pregnancy, which seriously endangers fetal health and still lacks effective therapeutic targets. Clostridium difficile (C. difficile) is associated with fetal birth weight, and its membrane vesicles (MVs) are pathogenic vectors. However, the role of C. difficile and its MVs in FGR remains unclear. Here we found that supplementation with C. difficile altered the characteristics of gut microbiota and reduced the birth weight in mice. Interestingly, C. difficile MVs entered placenta, inhibited trophoblast motility, and induced fetal weight loss in mice. Mechanistically, C. difficile MVs activated the PPAR pathway via enhancing the transcriptional activity of PPARγ promoter, consequently inhibiting trophoblast motility. Moreover, PPARγ expression was significantly elevated in FGR placenta, and negatively correlated with fetal birth weight. Together, our findings reveal the significance of C. difficile and its MVs in FGR, providing new insights into the mechanisms of FGR development.
Collapse
Affiliation(s)
- Zhiqiang Zha
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chunhong Jia
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ruisi Zhou
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qinlan Yin
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Hu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhipeng Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Linyu Peng
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yichi Zhang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaowei Qiu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yawen Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Wang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Menglan Pang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shijing Lu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chao Sheng
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Liping Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
23
|
Chen H, Shao LZ, Wang YX, Han ZJ, Wang YH, Li X, Chen JY, Liu TH. Causal Relationships Between Leukocyte Subsets and Adverse Fetal Outcomes: A Mendelian Randomization Study. Mediators Inflamm 2024; 2024:6349687. [PMID: 39748887 PMCID: PMC11695084 DOI: 10.1155/mi/6349687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/28/2024] [Accepted: 11/07/2024] [Indexed: 01/04/2025] Open
Abstract
Background: The tolerance and dynamic regulation of the maternal immune system during pregnancy are pivotal for ensuring fetal health. Immune cell subsets play a complex and crucial role in this process, closely linked to the neonatal health status. Despite recognizing the significance of dysregulation in the quantity and activity of immune cells in neonatal disease occurrence, their specific roles remain elusive, resulting in a dearth of clinically viable interventions for immune-mediated neonatal diseases. Materials and Methods: Employing two-sample Mendelian randomization (MR) methodology, this study systematically investigated 446 leukocyte features (N = 500,675), including leukocyte subsets, absolute cell (AC) counts, and morphological parameters (MP) and their correlation with seven adverse fetal outcomes (N = 1,100,458), encompassing fetal growth restriction (FGR), preterm birth (PTB), neonatal jaundice (NNJ), digestive system disorders of fetus and newborn (DSDFN), hemorrhagic and hematological disorders of fetus and newborn (HDFN), respiratory distress of newborn (RDN), and transitory disorders of metabolism specific to fetus and newborn (TDMSFN). Results: The results unveiled significant causal relationships between 301 leukocyte subsets and these seven adverse fetal outcomes, with 259, 245, 15, 44, 11, 32, and 68 pairs of notable associations for each adverse outcome, respectively. Furthermore, the study highlighted potential pathogenic mechanisms underlying the mutual influence among neonatal diseases. MR results indicated FGR as a robustly correlated risk factor for PTB and NNJ and showed a reciprocal causal relationship between NNJ and FGR. PTB exhibited a positive correlation with HDFN. Conclusions: This study provided profound insights into the intricate regulatory mechanisms of leukocyte subsets in neonatal diseases, paving the way for new avenues in the diagnosis and treatment of associated disorders.
Collapse
Affiliation(s)
- Hong Chen
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing 400016, China
| | - Li-Zhen Shao
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing 400016, China
| | - Ying-Xiong Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing 400016, China
| | - Zhi-Jie Han
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Yong-Heng Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing 400016, China
| | - Xia Li
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing 400016, China
| | - Jing-Yu Chen
- Department of Ultrasound, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Chongqing 400014, China
| | - Tai-Hang Liu
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
24
|
Hao H, Li F, Wang F, Ran J, Chen Y, Yang L, Ma H, Wang J, Yang H. Protective effect of metformin on the NG-nitro-l-arginine methyl ester (l-NAME)-induced rat models of preeclampsia. Biochem Biophys Res Commun 2024; 739:150996. [PMID: 39550868 DOI: 10.1016/j.bbrc.2024.150996] [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: 08/27/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Preeclampsia (PE) is a complex multi-organ disorder characterized by systemic inflammation, endothelial dysfunction, and vasoconstriction, which manifests as hypertension, with or without proteinuria. Effective preventive strategies for PE are currently lacking in clinical practice, leading to significant morbidity and mortality among mothers and newborns. OBJECTIVE This study aims to investigate the impact of metformin (MET) on the l-NAME-induced PE rat model, focusing on the mechanisms through which MET may exert its effects. METHODS Thirty pregnant Sprague-Dawley (SD) rats were randomly assigned to three groups: Control, PE, and PE + MET, on gestational day 0 (GD0). Regularly measure blood pressure and 24-h proteinuria, and collect tissue samples on GD20. Enzyme-linked immunosorbent assay (ELISA) was used to analyze inflammatory factors, endothelial function biomarkers, angiogenic factors, and apoptosis-related factors in the rat plasma. Western blot, RT-qPCR, and immunohistochemistry techniques were employed to determine the expression levels of key apoptotic proteins in placental tissue. RESULTS The study findings demonstrate that MET administration improves blood pressure and 24-h proteinuria, alleviates fetal growth restriction, ameliorate inflammation cytokines, and restores the balance of angiogenic factors and endothelial function. Moreover, MET inhibits the expression levels of critical apoptotic proteins in the plasma and placental tissue of PE-like rats. CONCLUSION The results suggest that MET shows promise in alleviating symptoms associated with l-NAME-induced PE in rats, preserving endothelial function, enhancing angiogenesis, reducing inflammation, inhibiting placental apoptosis, improving placental function, and promoting fetal growth. These findings highlight MET as a potential therapeutic agent for the prevention and treatment of preeclampsia.
Collapse
Affiliation(s)
- Huiniu Hao
- Department of Obstetrics, First Clinical College of Shanxi Medical University, Taiyuan, Shanxi Province, PR China
| | - Feng Li
- Department of Obstetrics, First Clinical College of Shanxi Medical University, Taiyuan, Shanxi Province, PR China
| | - Fang Wang
- Department of Obstetrics, First Clinical College of Shanxi Medical University, Taiyuan, Shanxi Province, PR China
| | - Jia Ran
- Department of Obstetrics, First Clinical College of Shanxi Medical University, Taiyuan, Shanxi Province, PR China
| | - Yinmin Chen
- Department of Obstetrics, First Clinical College of Shanxi Medical University, Taiyuan, Shanxi Province, PR China
| | - Lijun Yang
- Department of Obstetrics, First Clinical College of Shanxi Medical University, Taiyuan, Shanxi Province, PR China
| | - Huijing Ma
- Department of Obstetrics, First Clinical College of Shanxi Medical University, Taiyuan, Shanxi Province, PR China
| | - Jianli Wang
- Department of Obstetrics, First Clinical College of Shanxi Medical University, Taiyuan, Shanxi Province, PR China
| | - Hailan Yang
- Department of Obstetrics, First Clinical College of Shanxi Medical University, Taiyuan, Shanxi Province, PR China.
| |
Collapse
|
25
|
Davenport BN, Wilson RL, Williams AA, Jones HN. Placental nanoparticle-mediated IGF1 gene therapy corrects fetal growth restriction in a guinea pig model. Gene Ther 2024:10.1038/s41434-024-00508-3. [PMID: 39627510 DOI: 10.1038/s41434-024-00508-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 12/11/2024]
Abstract
Fetal growth restriction (FGR) caused by placental insufficiency is a major contributor to neonatal morbidity and mortality. There is currently no in utero treatment for placental insufficiency or FGR. The placenta serves as the vital communication, supply, exchange, and defense organ for the developing fetus and offers an excellent opportunity for therapeutic interventions. Here we show efficacy of repeated treatments of trophoblast-specific human insulin-like 1 growth factor (IGF1) gene therapy delivered in a non-viral, polymer nanoparticle to the placenta for the treatment of FGR. Using a guinea pig maternal nutrient restriction model (70% food intake) of FGR, nanoparticle-mediated IGF1 treatment was delivered to the placenta via ultrasound guidance across the second half of pregnancy, after establishment of FGR. This treatment resulted in correction of fetal weight in MNR + IGF1 animals compared to sham treated controls on an ad libitum diet, increased fetal blood glucose and decreased fetal blood cortisol levels compared to sham treated MNR, and showed no negative maternal side-effects. Overall, we show a therapy capable of positively impacting the entire pregnancy environment: maternal, placental, and fetal. This combined with our previous studies using this therapy at mid pregnancy in the guinea pig and in two different mouse model and three different human in vitro/ex vivo models, demonstrate the plausibility of this therapy for future human translation. Our overall goal is to improve health outcomes of neonates and decrease numerous morbidities associated with the developmental origins of disease.
Collapse
Affiliation(s)
- Baylea N Davenport
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Rebecca L Wilson
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, FL, USA.
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Alyssa A Williams
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Helen N Jones
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
26
|
Peng L, Yin Q, Wang X, Zhong Y, Wang Y, Cai W, Zhou R, Chen Y, Hu Y, Cheng Z, Jiang W, Yue X, Huang L. Pasteurized Akkermansia muciniphila Ameliorates Preeclampsia in Mice by Enhancing Gut Barrier Integrity, Improving Endothelial Function, and Modulating Gut Metabolic Dysregulation. Microorganisms 2024; 12:2483. [PMID: 39770686 PMCID: PMC11727688 DOI: 10.3390/microorganisms12122483] [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: 11/12/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 01/16/2025] Open
Abstract
Preeclampsia (PE) is a serious complication of pregnancy linked to endothelial dysfunction and an imbalance in the gut microbiota. While Akkermansia muciniphila (AKK) has shown promise in alleviating PE symptoms, the use of live bacteria raises safety concerns. This study explored the potential of pasteurized A. muciniphila (pAKK) as a safer alternative for treating PE, focusing on its effects on endothelial function and metabolic regulation. A PE mouse model was induced via the nitric oxide synthase inhibitor L-NAME, followed by treatment with either pAKK or live AKK. Fecal metabolomic profiling was performed via liquid chromatography-tandem mass spectrometry (LC-MS/MS), and in vivo and in vitro experiments were used to assess the effects of pAKK on endothelial function and metabolic pathways. pAKK exhibited therapeutic effects comparable to those of live AKK in improving L-NAME-induced PE-like phenotypes in mice, including enhanced gut barrier function and reduced endotoxemia. pAKK also promoted placental angiogenesis by restoring endothelial nitric oxide synthase (eNOS) activity and nitric oxide (NO) production. The in vitro experiments further confirmed that pAKK alleviated L-NAME-induced NO reduction and endothelial dysfunction in human umbilical vein endothelial cells (HUVECs). Metabolomic analysis revealed that both pAKK and live AKK reversed metabolic disturbances in PE by modulating key metabolites and pathways related to unsaturated fatty acid biosynthesis, folate, and linoleic acid metabolism. As a postbiotic, pAKK may support existing treatments for preeclampsia by improving gut barrier function, restoring endothelial function, and regulating metabolic dysregulation, offering a safer alternative to live bacteria. These findings highlight the potential clinical value of pAKK as an adjunctive therapy in managing PE.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Xiaojing Yue
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; (L.P.)
| | - Liping Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; (L.P.)
| |
Collapse
|
27
|
Givens M, Valcheva I, Einerson BD, Rogozińska E, Jauniaux E. Evaluation of maternal serum protein biomarkers in the prenatal evaluation of placenta accreta spectrum: A systematic scoping review. Acta Obstet Gynecol Scand 2024; 103:2335-2347. [PMID: 39004916 PMCID: PMC11610010 DOI: 10.1111/aogs.14918] [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: 04/11/2024] [Revised: 05/29/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024]
Abstract
INTRODUCTION Placenta accreta spectrum (PAS) is an increasingly commonly reported condition due to the continuous increase in the rate of cesarean deliveries (CD) worldwide; however, the prenatal screening for pregnant patients at risk of PAS at birth remains limited, in particular when imaging expertise is not available. MATERIAL AND METHODS Two major electronic databases (MEDLINE and Embase) were searched electronically for articles published in English between October 1992 and January 2023 using combinations of the relevant medical subject heading terms and keywords. Two independent reviewers selected observational studies that provided data on one or more measurement of maternal blood-specific biomarker(s) during pregnancies with PAS at birth. PRISMA Extension for Scoping Review (PRISMA-ScR) was used to extract data and report results. RESULTS Of the 441 reviewed articles, 29 met the inclusion criteria reporting on 34 different biomarkers. 14 studies were retrospective and 15 prospective overall including 18 251 participants. Six studies had a cohort design and the remaining a case-control design. Wide clinical heterogeneity was found in the included studies. In eight studies, the samples were obtained in the first trimester; in five, the samples were collected on hospital admission for delivery; and in the rest, the samples were collected during the second and/or third trimester. CONCLUSIONS Measurements of serum biomarkers, some of which have been or are still used in screening for other pregnancy complications, could contribute to the prenatal evaluation of patients at risk of PAS at delivery; however, important evidence gaps were identified for suitable cutoffs for most biomarkers, variability of gestational age at sampling and the potential overlap of the marker values with other placental-related complications of pregnancy.
Collapse
Affiliation(s)
- Matthew Givens
- Department of Obstetrics and Gynecology (Drs Givens and Einerson)University of Utah Health (UUH)Salt Lake CityUtahUSA
| | - Ivaila Valcheva
- EGA Institute for Women's Health, Faculty of Population Health SciencesUniversity College LondonLondonUK
| | - Brett D. Einerson
- Department of Obstetrics and Gynecology (Drs Givens and Einerson)University of Utah Health (UUH)Salt Lake CityUtahUSA
| | - Ewelina Rogozińska
- The EVIdencE Synthesis and Methodology Group for Women's Health Research (EVIE)Institute of Clinical Trials & Methodology, University College LondonLondonUK
| | - Eric Jauniaux
- EGA Institute for Women's Health, Faculty of Population Health SciencesUniversity College LondonLondonUK
| |
Collapse
|
28
|
Hennessy A, Heffernan S, Pears S, Roshan N, Romano AB, Karumanchi A, Hyett J, Waugh R, Iliopoulos J, Makris A. Birthweight in a non-human primate model of placental ischaemia. Pregnancy Hypertens 2024; 38:101160. [PMID: 39461249 DOI: 10.1016/j.preghy.2024.101160] [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: 05/12/2024] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 10/29/2024]
Abstract
Animal models for preeclampsia are mostly determined by the experimental induction of hypertension, proteinuria and latterly, endogenous production of anti-angiogenic factors (sFlt-1). The focus on maternal outcome measures is more immediately obvious, with comparative and sequential data of blood pressure and urine protein excretion. In non-human primates, the data concerning birthweight requires a greater number of observations and thus will be accumulated over a longer period of time and a greater number of experimental protocols. The following represents the outcome of over 20 years of experimental preeclampsia (EPE) compared with normal pregnancy outcome data in baboons. MethodsThis data represents the outcomes from 91 pregnancies over the last 25 years at the Australian National Baboon Colony. These pregnancies are attributed to females who had experimental preeclampsia (EPE) and those within the general colony. EPE was induced at day 130 (of 182 days gestation length), and in some protocols, treatments such as inhibitory RNA or placental growth factor (PlGF) were tested. All studies were approved by the institutional Animal Welfare Committee. RESULTS: The overall neonatal birthweight was 697 g ± 115 g. The average birthweight for normal males was 770 ± 105 g; and for male offspring of animals with EPE, 680 ± 113 g; for normal females was 640 ± 95 g and females from EPE pregnancies, 690 ± 43 g. There was only a significant difference in weight for females compared to males overall (p = 0.002), and there was no significant difference in birthweight for males or females subjected to EPE. Correction for treated EPE did not change the outcome. CONCLUSIONS: These data indicate that in a non-human primate model of placental dysfunction through late pregnancy acute ischaemia, there is no measurable effect on baby birthweight compared to normal pregnancy, and no impact from a number of current experimental treatment strategies.
Collapse
Affiliation(s)
- A Hennessy
- Sydney Local health District, Sydney, Australia; South Western Sydney Local Health District, Sydney, Australia; Western Sydney University, Sydney, Australia.
| | - S Heffernan
- Sydney Local health District, Sydney, Australia
| | - S Pears
- Sydney Local health District, Sydney, Australia
| | - N Roshan
- South Western Sydney Local Health District, Sydney, Australia
| | - A B Romano
- Sydney Local health District, Sydney, Australia
| | - A Karumanchi
- Cedars Sinai Medical Centre, Los Angeles, United States
| | - J Hyett
- South Western Sydney Local Health District, Sydney, Australia
| | - R Waugh
- Sydney Local health District, Sydney, Australia
| | - J Iliopoulos
- South Western Sydney Local Health District, Sydney, Australia; Western Sydney University, Sydney, Australia
| | - A Makris
- South Western Sydney Local Health District, Sydney, Australia; Western Sydney University, Sydney, Australia
| |
Collapse
|
29
|
Nurkkala J, Vaura F, Toivonen J, Niiranen T. Genetics of hypertension-related sex differences and hypertensive disorders of pregnancy. Blood Press 2024; 33:2408574. [PMID: 39371034 DOI: 10.1080/08037051.2024.2408574] [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: 06/21/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024]
Abstract
Background: Hypertension and hypertensive disorders of pregnancy (HDP) cause a significant burden of disease on societies and individuals by increasing cardiovascular disease risk. Environmental risk factors alone do not explain the observed sexual dimorphism in lifetime blood pressure (BP) trajectories nor inter-individual variation in HDP risk. Methods: In this short review, we focus on the genetics of hypertension-related sex differences and HDP and discuss the importance of genetics utilization for sex-specific hypertension risk prediction. Results: Population and twin studies estimate that 28-66% of variation in BP levels and HDP is explained by genetic variation, while genomic wide association studies suggest that BP traits and HDP partly share a common genetic background. Moreover, environmental and epigenetic regulation of these genes differ by sex and oestrogen receptors in particular are shown to convey cardio- and vasculoprotective effects through epigenetic regulation of DNA. The majority of known genetic variation in hypertension and HDP is polygenic. Polygenic risk scores for BP display stronger associations with hypertension risk in women than in men and are associated with sex-specific age of hypertension onset. Monogenic forms of hypertension are rare and mostly present equally in both sexes. Conclusion: Despite recent genetic discoveries providing new insights into HDP and sex differences in BP traits, further research is needed to elucidate the underlying biology. Emphasis should be placed on demonstrating the added clinical value of these genetic discoveries, which may eventually facilitate genomics-based personalized treatments for hypertension and HDP.
Collapse
Affiliation(s)
- Jouko Nurkkala
- Division of Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital, Turku, Finland
- Department of Anesthesiology and Intensive Care, University of Turku, Turku, Finland
| | - Felix Vaura
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Jenni Toivonen
- Division of Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital, Turku, Finland
- Department of Anesthesiology and Intensive Care, University of Turku, Turku, Finland
| | - Teemu Niiranen
- Department of Internal Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Turku, Finland
| |
Collapse
|
30
|
Nedberg NH, Nystad M, Ahlen MT, Bertelsen EL, Guz K, Uhrynowska M, Dębska M, Gierszon A, Orzińska A, Husebekk A, Brojer E, Staff AC, Tiller H. Placenta-associated biomarkers and pregnancy outcome in HPA-1a alloimmunization: A prospective cohort study. Placenta 2024; 158:185-191. [PMID: 39486121 DOI: 10.1016/j.placenta.2024.10.014] [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: 07/14/2024] [Revised: 10/11/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024]
Abstract
INTRODUCTION Fetal and neonatal alloimmune thrombocytopenia (FNAIT) results from parental incompatibility in human platelet antigens (HPA) and subsequent maternal sensitization. The HPA-1a epitope is also expressed on placental tissue. Chronic placental inflammation and lower birth weight is observed more often in HPA-1a alloimmunized pregnancies, suggesting a placental component in the pathophysiology of FNAIT. Today, prediction of FNAIT severity is limited. The aim of the study was to investigate whether dysregulated maternal angiogenic proteins are associated with neonatal outcome in HPA-1a alloimmunized pregnancies. MATERIAL AND METHODS Eighty-seven HPA-1a negative pregnant women were identified from a large prospective screening study in Poland (PREVFNAIT) including 43 HPA-1a immunized and 44 non-immunized controls. Placental growth factor (PlGF), soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin (sEng) were measured in maternal plasma from 2nd and 3rd trimester by enzyme-linked immunosorbent assay and levels/ratios were compared between study groups, using uni- and multivariable analyses. Main outcome measures were either classic FNAIT-related (severe thrombocytopenia, petechia, intracranial hemorrhage), placenta-related (small for gestational age) or a composite variable combining them all. RESULTS There were no significant differences in plasma concentrations of sFlt-1, PlGF, sEng nor sFlt-1/PlGF ratio when comparing immunized and non-immunized pregnancies. Among HPA-1a alloimmunized pregnancies, increasing levels of the sFlt-1 protein in 3rd trimester were significantly associated with lower neonatal platelet count (multivariable linear regression, p = 0.024). Increased sFlt-1 and sFlt-1/PlGF ratio in 3rd trimester were significantly associated with higher odds of a composite adverse neonatal outcome in alloimmunized pregnancies (multivariable logistic regression, p = 0.029 and p = 0.019, respectively). CONCLUSION An anti-angiogenic profile in HPA-1a alloimmunized mothers is associated with a composite adverse neonatal outcome. This suggests that sFlt-1 and the sFlt-1/PlGF ratio may assist in predelivery risk stratification and clinical management decisions for FNAIT.
Collapse
Affiliation(s)
- Nora Hersoug Nedberg
- Immunology Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, Tromsø, Norway; Women's Health and Perinatology Research Group, Department of Clinical Medicine, Faculty of Health Sciences, UiT the Arctic University of Norway, Tromsø, Norway
| | - Mona Nystad
- Women's Health and Perinatology Research Group, Department of Clinical Medicine, Faculty of Health Sciences, UiT the Arctic University of Norway, Tromsø, Norway; Department of Obstetrics and Gynecology, University Hospital of North Norway, Tromsø, Norway
| | - Maria Therese Ahlen
- Immunology Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, Tromsø, Norway; Norwegian National Unit for Platelet Immunology, Department of Laboratory Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Eirin Listau Bertelsen
- Immunology Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, Tromsø, Norway
| | - Katarzyna Guz
- Department of Hematological and Transfusion Immunology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Małgorzata Uhrynowska
- Department of Hematological and Transfusion Immunology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Marzena Dębska
- Department of Obstetrics and Perinatology, National Medical Institute of the Ministry of Interior and Administration, Warsaw, Poland
| | - Agnieszka Gierszon
- Department of Hematological and Transfusion Immunology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Agnieszka Orzińska
- Department of Hematological and Transfusion Immunology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Anne Husebekk
- Immunology Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, Tromsø, Norway
| | - Ewa Brojer
- Department of Hematological and Transfusion Immunology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Anne Cathrine Staff
- Division of Obstetrics and Gynecology, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Heidi Tiller
- Women's Health and Perinatology Research Group, Department of Clinical Medicine, Faculty of Health Sciences, UiT the Arctic University of Norway, Tromsø, Norway; Department of Obstetrics and Gynecology, University Hospital of North Norway, Tromsø, Norway.
| |
Collapse
|
31
|
Ricart M, Bonacina E, Garcia‐Manau P, López M, Caamiña S, Vives À, Lopez‐Quesada E, Maroto A, de Mingo L, Pintado E, Ferrer‐Costa R, Martín L, Rodriguez‐Zurita A, Garcia E, Pallarols M, Pratcorona L, Teixidor M, Orizales‐Lago C, Ocaña V, del Barco E, Carreras E, Suy A, Mendoza M. Placental growth factor at 24-28 weeks for aspirin discontinuation in pregnancies at high risk for preterm preeclampsia: Post hoc analysis of StopPRE trial. Acta Obstet Gynecol Scand 2024; 103:2273-2280. [PMID: 39171611 PMCID: PMC11502455 DOI: 10.1111/aogs.14955] [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: 05/10/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024]
Abstract
INTRODUCTION This study aims to evaluate the safety of discontinuing aspirin treatment at 24-28 weeks in women at high risk after first-trimester combined screening for preeclampsia (PE) and normal placental growth factor (PlGF) levels at 24-28 weeks of gestation. MATERIAL AND METHODS This is a post hoc analysis of the StopPRE trial, conducted at nine Spanish maternity hospitals from September 2019 to September 2021. In the StopPRE trial, all high-risk single pregnancies identified during first-trimester screening for PE were treated with 150 mg of daily aspirin. Out of 1604 eligible women with a soluble fms-like tyrosine kinase-1 to PlGF ratio (sFlt-1/PlGF) ≤38 at 24-28 weeks, 968 were randomly assigned in a 1:1 ratio to either continue aspirin until 36 weeks (control group) or discontinue it (intervention group). In this secondary analysis, only women with PlGF ≥100 pg/mL at 24-28 weeks were included. As in the StopPRE trial, the non-inferiority margin was set at a 1.9% difference in preterm PE incidence between the groups. RESULTS Among the 13 983 screened pregnant women, 1984 (14.2%) were deemed high-risk for preterm PE, of which 397 (20.0%) were ineligible, 636 declined participation, and 32 were excluded. Ultimately, 919 women with PlGF >100 pg/mL were randomized and included in this analysis. Preterm PE occurred in 0.9% of the intervention group (4 out of 465) and 1.5% of the control group (7 out of 454), indicating non-inferiority of aspirin discontinuation. There were no significant differences between the groups in adverse pregnancy outcomes before 37 weeks, at <34 weeks, or ≥37 weeks. Minor antepartum hemorrhage incidence was significantly lower in the intervention group (absolute difference, -5.96; 95% CI, -10.10 to -1.82). CONCLUSIONS Discontinuation of aspirin treatment at 24-28 weeks in women with PlGF levels ≥100 pg/mL was non-inferior to continuing until 36 weeks for preventing preterm PE. However, these findings should be interpreted with caution, as they originate from a subanalysis of the StopPRE trial.
Collapse
Affiliation(s)
- Marta Ricart
- Department of Pediatrics, Obstetrics and Gynecology, and Preventive Medicine and Public HealthUniversitat Autònoma de BarcelonaBellaterraSpain
- Department of ObstetricsHospital Universitari Germans Trias i PujolBadalonaSpain
| | - Erika Bonacina
- Maternal Fetal Medicine Unit, Department of ObstetricsVall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| | - Pablo Garcia‐Manau
- Maternal Fetal Medicine Unit, Department of ObstetricsVall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| | - Monica López
- Department of ObstetricsHospital Universitari de Tarragona Joan XXIIITarragonaSpain
| | - Sara Caamiña
- Department of ObstetricsHospital Universitario Nuestra Señora de CandelariaSanta Cruz de TenerifeSpain
| | - Àngels Vives
- Department of ObstetricsConsorci Sanitari de TerrassaTerrassaSpain
| | - Eva Lopez‐Quesada
- Department of ObstetricsHospital Universitari Mútua TerrassaTerrassaSpain
| | - Anna Maroto
- Department of ObstetricsHospital Universitari de Girona Doctor Josep TruetaGironaSpain
| | - Laura de Mingo
- Department of ObstetricsHospital Universitario Severo OchoaLeganésSpain
| | - Elena Pintado
- Department of ObstetricsHospital Universitario de GetafeGetafeSpain
| | - Roser Ferrer‐Costa
- Department of BiochemistryVall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| | - Lourdes Martín
- Department of ObstetricsHospital Universitari de Tarragona Joan XXIIITarragonaSpain
| | - Alicia Rodriguez‐Zurita
- Department of ObstetricsHospital Universitario Nuestra Señora de CandelariaSanta Cruz de TenerifeSpain
| | - Esperanza Garcia
- Department of ObstetricsConsorci Sanitari de TerrassaTerrassaSpain
| | - Mar Pallarols
- Department of ObstetricsHospital Universitari Mútua TerrassaTerrassaSpain
| | - Laia Pratcorona
- Department of Pediatrics, Obstetrics and Gynecology, and Preventive Medicine and Public HealthUniversitat Autònoma de BarcelonaBellaterraSpain
- Department of ObstetricsHospital Universitari Germans Trias i PujolBadalonaSpain
| | - Mireia Teixidor
- Department of ObstetricsHospital Universitari de Girona Doctor Josep TruetaGironaSpain
| | | | - Vanesa Ocaña
- Department of ObstetricsHospital Universitario de GetafeGetafeSpain
| | - Esther del Barco
- Maternal Fetal Medicine Unit, Department of ObstetricsVall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| | - Elena Carreras
- Maternal Fetal Medicine Unit, Department of ObstetricsVall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| | - Anna Suy
- Department of Pediatrics, Obstetrics and Gynecology, and Preventive Medicine and Public HealthUniversitat Autònoma de BarcelonaBellaterraSpain
- Maternal Fetal Medicine Unit, Department of ObstetricsVall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| | - Manel Mendoza
- Department of Pediatrics, Obstetrics and Gynecology, and Preventive Medicine and Public HealthUniversitat Autònoma de BarcelonaBellaterraSpain
- Maternal Fetal Medicine Unit, Department of ObstetricsVall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| |
Collapse
|
32
|
Melamed N, Kingdom JC, Fu L, Yip PM, Arruda-Caycho I, Hui D, Hladunewich MA. Predictive and Diagnostic Value of the Angiogenic Proteins in Patients With Chronic Kidney Disease. Hypertension 2024; 81:2251-2262. [PMID: 39162032 DOI: 10.1161/hypertensionaha.124.23411] [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: 05/23/2024] [Accepted: 07/30/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Our objective was to investigate the predictive and diagnostic accuracy of the angiogenic proteins sFlt-1 (soluble fms-like tyrosine kinase-1) and PlGF (placental growth factor) for preterm preeclampsia and explore the relationship between renal function and these proteins. METHODS We completed a blinded, prospective, longitudinal, observational study of patients with chronic kidney disease followed at a tertiary center (2018-2023). Serum samples were obtained at 3 time points along gestation (planned sampling): 12-16, 18-22, and 28-32 weeks. In addition, samples were obtained whenever preeclampsia was suspected (indicated sampling). sFlt-1 and PlGF levels remained concealed until the study ended. The primary outcome was preterm preeclampsia. The planned and indicated samples were used to estimate the predictive and diagnostic accuracy of the angiogenic proteins, respectively. RESULTS Of the 97 participants, 21 (21.6%) experienced preterm preeclampsia. In asymptomatic patients with chronic kidney disease, the angiogenic proteins were predictive of preterm preeclampsia only when sampled in the third trimester, in which case the sFlt-1/PlGF ratio (false positive rate of 37% for a detection rate of 80%) was more predictive than either sFlt-1 or PlGF in isolation. In patients with suspected preeclampsia, the diagnostic accuracy of the sFlt-1/PlGF ratio (false positive rate of 26% for a detection rate of 80%) was higher than that of sFlt-1 and PlGF in isolation. Diminished renal function was associated with increased levels of PlGF. CONCLUSIONS sFlt-1 and PlGF can effectively predict and improve the diagnostic accuracy for preterm preeclampsia among patients with chronic kidney disease. The optimal sFlt-1/PlGF ratio cutoff to rule out preeclampsia may need to be lower in patients with impaired renal function.
Collapse
Affiliation(s)
- Nir Melamed
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, Temerty Faculty of Medicine (N.M., I.A.-C., D.H.), University of Toronto, Ontario, Canada
| | - John C Kingdom
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Mount Sinai Hospital, Temerty Faculty of Medicine (J.C.K.), University of Toronto, Ontario, Canada
| | - Lei Fu
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre (L.F., P.M.Y.), University of Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology (L.F., P.M.Y.), University of Toronto, Ontario, Canada
| | - Paul M Yip
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre (L.F., P.M.Y.), University of Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology (L.F., P.M.Y.), University of Toronto, Ontario, Canada
| | - Isabel Arruda-Caycho
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, Temerty Faculty of Medicine (N.M., I.A.-C., D.H.), University of Toronto, Ontario, Canada
| | - Dini Hui
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, Temerty Faculty of Medicine (N.M., I.A.-C., D.H.), University of Toronto, Ontario, Canada
| | - Michelle A Hladunewich
- Division of Nephrology and Obstetric Medicine, Department of Medicine, Sunnybrook Health Sciences Centre, Temerty Faculty of Medicine (M.A.H.), University of Toronto, Ontario, Canada
| |
Collapse
|
33
|
Zhang F, Wang H. Effect of low-dose aspirin intervention on pre-eclampsia prevention in high-risk pregnant women and its impact on postpartum hemorrhage. Front Med (Lausanne) 2024; 11:1414697. [PMID: 39526246 PMCID: PMC11543432 DOI: 10.3389/fmed.2024.1414697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
Background Pre-eclampsia, characterized by hypertension and organ dysfunction during pregnancy, poses significant risks to both maternal and fetal health. Aspirin, known for its antiplatelet properties, has been extensively utilized to mitigate pregnancy-related complications. However, the efficacy of low-dose aspirin in managing pre-eclampsia among high-risk pregnant women and its potential impact on postpartum hemorrhage remain contentious topics. Methods A retrospective analysis was conducted on 344 pregnant women diagnosed with high-risk factors for pre-eclampsia. Among them, 152 received intervention with low-dose aspirin, while the rest did not receive it. The incidence of pre-eclampsia, as well as related complications and outcomes associated with bleeding, were compared and evaluated between the two groups. Results The study findings indicate a significant reduction in the incidence of pre-eclampsia among pregnant women receiving low-dose aspirin intervention, along with a significantly reduced risk of complications. Additionally, there was no significant statistical difference in postpartum hemorrhage between the two groups (p > 0.05). The safety profile of aspirin usage was found to be favorable. Conclusion Low-dose aspirin demonstrates promising efficacy as an intervention strategy for high-risk preeclamptic women. It does not increase the risk of postpartum hemorrhage and reduces the occurrence of complications associated with preeclampsia. Therefore, low-dose aspirin presents a potential preventive measure against adverse outcomes associated with high-risk pregnancies related to preeclampsia. Further research is necessary to validate and elucidate the optimal dosage and timing of administration for maximal benefits.
Collapse
Affiliation(s)
| | - Huijuan Wang
- Department of Obstetrics and Gynecology, Xi’an People's Hospital (Xi’an Fourth Hospital), Xi’an, Shaanxi, China
| |
Collapse
|
34
|
Hong J, Crawford K, Daly M, Clifton V, da Silva Costa F, Perkins AV, Matsika A, Lourie R, Kumar S. Utility of placental biomarkers and fetoplacental Dopplers in predicting likely placental pathology in early and late fetal growth restriction - A prospective study. Placenta 2024; 156:20-29. [PMID: 39232442 DOI: 10.1016/j.placenta.2024.08.016] [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: 07/05/2024] [Revised: 08/07/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024]
Abstract
INTRODUCTION The aim of this study was to evaluate the association between placental abnormalities, placental biomarkers, and fetoplacental Dopplers in a cohort of pregnancies complicated by fetal growth restriction (FGR). We also ascertained the risk of perinatal mortality, severe neurological morbidity, and severe non-neurological morbidity by type of placental abnormality. METHODS This was a prospective cohort study. Multivariable logistic regression was used to evaluate the effect of early vs. late FGR, placental biomarkers and fetoplacental Dopplers on Maternal Vascular Malperfusion (MVM) which was the commonest placental abnormality identified. RESULTS There were 161 (53.5 %) early FGR and 140 (46.5 %) late FGR cases. MVM abnormalities were present in 154 (51.2 %), VUE in 45 (14.6 %), FVM in 16 (5.3 %), DVM in 14 (4.7 %) and CHI in 4 (1.3 %) cases. The odds of MVM were higher in early compared to late FGR cohort (OR 1.89, 95%CI 1.14, 3.14, p = 0.01). Low maternal PlGF levels <100 ng/L (OR 2.34, 95%CI 1.27,4.31, p = 0.01), high sFlt-1 level (OR 2.13, 95%CI 1.35, 3.36, p = 0.001) or elevated sFlt-1/PlGF ratio (OR 3.48, 95%CI 1.36, 8.91, p = 0.01) were all associated with MVM. Increased UA PI > 95th centile (OR 2.91, 95%CI 1.71, 4.95, p=<0.001) and mean UtA PI z-score (OR 1.74, 95%CI 1.15, 2.64, p = 0.01) were associated with higher odds of MVM. Rates of severe non-neurological morbidity were highest in the MVM, FVM, and CHI cohorts (44.8 %, 50 %, and 50 % respectively). CONCLUSION MVM was the commonest placental abnormality in FGR, particularly in early-onset disease. Low maternal PlGF levels, high sFlt-1 levels, elevated sFlt-1/PlGF ratio, and abnormal fetoplacental Dopplers were also significantly associated with MVM. MVM, FVM, and CHI abnormalities were associated with lower median birthweight, higher rates of preterm birth, operative birth for non-reassuring fetal status, and severe neonatal non-neurological morbidity.
Collapse
Affiliation(s)
- Jesrine Hong
- Mater Research Institute, University of Queensland, Level 3, Aubigny Place, Raymond Terrace, South Brisbane, Queensland, 4101, Australia; School of Medicine, The University of Queensland, Herston, Queensland, 4006, Australia; Department of Obstetrics and Gynecology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Kylie Crawford
- Mater Research Institute, University of Queensland, Level 3, Aubigny Place, Raymond Terrace, South Brisbane, Queensland, 4101, Australia; School of Medicine, The University of Queensland, Herston, Queensland, 4006, Australia
| | - Matthew Daly
- Mater Research Institute, University of Queensland, Level 3, Aubigny Place, Raymond Terrace, South Brisbane, Queensland, 4101, Australia; School of Medicine, The University of Queensland, Herston, Queensland, 4006, Australia
| | - Vicki Clifton
- Mater Research Institute, University of Queensland, Level 3, Aubigny Place, Raymond Terrace, South Brisbane, Queensland, 4101, Australia
| | - Fabricio da Silva Costa
- School of Medicine and Dentistry, Griffith University and Maternal Fetal Medicine Unit, Gold Coast University Hospital, Gold Coast, Queensland, Australia
| | - Anthony V Perkins
- School of Health, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - Admire Matsika
- Mater Research Institute, University of Queensland, Level 3, Aubigny Place, Raymond Terrace, South Brisbane, Queensland, 4101, Australia
| | - Rohan Lourie
- Mater Research Institute, University of Queensland, Level 3, Aubigny Place, Raymond Terrace, South Brisbane, Queensland, 4101, Australia
| | - Sailesh Kumar
- Mater Research Institute, University of Queensland, Level 3, Aubigny Place, Raymond Terrace, South Brisbane, Queensland, 4101, Australia; School of Medicine, The University of Queensland, Herston, Queensland, 4006, Australia; NHMRC Centre for Research Excellence in Stillbirth, Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
35
|
Davenport B, Wilson R, Williams A, Jones H. Placental Nanoparticle-mediated IGF1 Gene Therapy Corrects Fetal Growth Restriction in a Guinea Pig Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.587765. [PMID: 38645174 PMCID: PMC11030242 DOI: 10.1101/2024.04.05.587765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Fetal growth restriction (FGR) caused by placental insufficiency is a major contributor to neonatal morbidity and mortality. There is currently no in utero treatment for placental insufficiency or FGR. The placenta serves as the vital communication, supply, exchange, and defense organ for the developing fetus and offers an excellent opportunity for therapeutic interventions. Here we show efficacy of repeated treatments of trophoblast-specific human insulin-like 1 growth factor (IGF1) gene therapy delivered in a non-viral, polymer nanoparticle to the placenta for the treatment of FGR. Using a guinea pig maternal nutrient restriction model (70% food intake) of FGR, nanoparticle-mediated IGF1 treatment was delivered to the placenta via ultrasound guidance across the second half of pregnancy, after establishment of FGR. This treatment resulted in correction of fetal weight in MNR + IGF1 animals compared to sham treated controls on an ad libitum diet, increased fetal blood glucose and decreased fetal blood cortisol levels compared to sham treated MNR, and showed no negative maternal side-effects. Overall, we show a therapy capable of positively impacting the entire pregnancy environment: maternal, placental, and fetal. This combined with our previous studies using this therapy at mid pregnancy in the guinea pig and in two different mouse model and three different human in vitro/ex vivo models, demonstrate the plausibility of this therapy for future human translation. Our overall goal is to improve health outcomes of neonates and decrease numerous morbidities associated with the developmental origins of disease.
Collapse
|
36
|
Sun H, Jiao J, Wang Y, Zhu C, Wang S, Wang Y, Ban B, Guo Y, Ren Y. Ultrasound based radiomics model for assessment of placental function in pregnancies with preeclampsia. Sci Rep 2024; 14:21123. [PMID: 39256496 PMCID: PMC11387498 DOI: 10.1038/s41598-024-72046-2] [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: 12/20/2023] [Accepted: 09/03/2024] [Indexed: 09/12/2024] Open
Abstract
The goal of our research is to elucidate and better assess placental function in rats with preeclampsia through an innovative application of ultrasound-based radiomics. Using a rat model induced with L-NAME, we carefully investigated placental dysfunction via microstructural analysis and immunoprotein level assessment. Employing the Boruta feature selection method on ultrasound images facilitated the identification of crucial features, consequently enabling the development of a robust model for classifying placental dysfunction. Our study included 12 pregnant rats, and thorough placental evaluations were conducted on 160 fetal rats. Distinct alterations in placental microstructure and angiogenic factor expression were evident in rats with preeclampsia. Leveraging high-throughput mining of quantitative image features, we extracted 558 radiomic features, which were subsequently used to construct an impressive evaluation model with an area under the receiver operating curve (AUC) of 0.95. This model also exhibited a remarkable sensitivity, specificity, accuracy, positive predictive value, and negative predictive value of 88.7%, 91.5%, 90.2%, 90.4%, and 90.0%, respectively. Our findings highlight the ability of ultrasound-based radiomics to detect abnormal placental features, demonstrating its potential for evaluating both normative and impaired placental function with high precision and reliability.
Collapse
Affiliation(s)
- Hongshuang Sun
- Department of Ultrasound Medicine, Affiliated Hospital of Jining Medical College, Shandong, 272029, China
- Key Laboratory for Experimental Teratology of the Ministry of Education and Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Jing Jiao
- Department of Electronic Engineering, School of Information Science and Technology, Fudan University, Handan Road, Yangpu District, Shanghai, 200433, China
- The Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, 200433, China
| | - Yicong Wang
- Medical Imaging Department, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong, China
| | - Chen Zhu
- Department of Ultrasound Medicine, Obstetrics and Gynecology Hospital of Fudan University, No. 128, Shenyang Road, Shanghai, 200090, China
| | - Shaochun Wang
- Department of Ultrasound Medicine, Affiliated Hospital of Jining Medical College, Shandong, 272029, China
- Key Laboratory for Experimental Teratology of the Ministry of Education and Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yuanyuan Wang
- Department of Electronic Engineering, School of Information Science and Technology, Fudan University, Handan Road, Yangpu District, Shanghai, 200433, China
- The Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, 200433, China
| | - Bo Ban
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong, China.
| | - Yi Guo
- Department of Electronic Engineering, School of Information Science and Technology, Fudan University, Handan Road, Yangpu District, Shanghai, 200433, China.
- The Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, 200433, China.
| | - Yunyun Ren
- Department of Ultrasound Medicine, Obstetrics and Gynecology Hospital of Fudan University, No. 128, Shenyang Road, Shanghai, 200090, China.
| |
Collapse
|
37
|
Nema J, Sundrani D, Randhir K, Deshpande J, Lalwani S, Wagh G, Gupte S, Joshi S. Maternal angiogenic factor disruptions prior to clinical diagnosis of preeclampsia: insights from the REVAMP study. Hypertens Res 2024; 47:2532-2548. [PMID: 38965425 DOI: 10.1038/s41440-024-01775-8] [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: 12/12/2023] [Revised: 05/12/2024] [Accepted: 06/06/2024] [Indexed: 07/06/2024]
Abstract
Preeclampsia is characterized by impaired angiogenesis and assessment of angiogenic factors can play a crucial role in the early diagnosis of preeclampsia. The current study reports the levels of angiogenic factors longitudinally from early pregnancy in women with preeclampsia and in the subtypes of preeclampsia, to identify their role in early prediction of preeclampsia. A total of 1154 women with singleton pregnancies were recruited in early pregnancy from 2 hospitals. Blood samples were collected, plasma samples were separated and stored at four time points across gestation: V1 = 11-14 weeks, V2 = 18-22 weeks, V3 = 26-28 weeks, and V4 = at delivery. The current study includes a total of 108 women developed preeclampsia (PE), and 216 matched controls. Angiogenic factors were estimated using commercially available ELISA kits. Receiver operating characteristic (ROC) curves were used to evaluate the potential diagnostic value in the prediction of PE. Lower levels of VEGF, PlGF, and higher levels of sEng and sEng/PlGF ratio (p < 0.05 for all) predate clinical diagnosis in women with preeclampsia. sEng levels and sEng/PlGF ratio showed significant correlation with odds of preeclampsia at all the timepoints. This study identifies a cut off of 33.5 for sFlt-1/PlGF and 25.9 for sEng/PlGF for prediction of early onset preeclampsia. This study reports various angiogenic factors serially across gestation in a general population to identify women at risk of developing preeclampsia and its subtypes. The study also reports a potential biomarker and a pragmatic window for estimation of angiogenic markers to identify women at risk.
Collapse
Affiliation(s)
- Juhi Nema
- Mother and Child Health, ICMR- Collaborating Centre of Excellence (ICMR-CCoE), Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune Satara Road, Pune, 411043, India
| | - Deepali Sundrani
- Mother and Child Health, ICMR- Collaborating Centre of Excellence (ICMR-CCoE), Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune Satara Road, Pune, 411043, India
| | - Karuna Randhir
- Mother and Child Health, ICMR- Collaborating Centre of Excellence (ICMR-CCoE), Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune Satara Road, Pune, 411043, India
| | - Juilee Deshpande
- Mother and Child Health, ICMR- Collaborating Centre of Excellence (ICMR-CCoE), Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune Satara Road, Pune, 411043, India
| | - Sanjay Lalwani
- Department of Pediatrics, Bharati Medical College and Hospital, Bharati Vidyapeeth (Deemed to be University), Pune, 411043, India
| | - Girija Wagh
- Department of Obstetrics and Gynaecology, Bharati Medical College and Hospital, Bharati Vidyapeeth (Deemed to be University), Pune, 411043, India
| | - Sanjay Gupte
- Gupte Hospital and Research Centre, Pune, 411004, India
| | - Sadhana Joshi
- Mother and Child Health, ICMR- Collaborating Centre of Excellence (ICMR-CCoE), Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune Satara Road, Pune, 411043, India.
| |
Collapse
|
38
|
Czeiger S, Weissbach T, Zloto K, Wiener A, Nir O, Massarwa A, Weisz B, Bartal MF, Ulman RY, Bart Y, Achiron R, Kivilevitch Z, Mazaki-Tovi S, Kassif E. Umbilical-portal-systemic venous shunt and intrauterine growth restriction: an inquiry from a prospective study. Am J Obstet Gynecol 2024; 231:340.e1-340.e16. [PMID: 38218510 DOI: 10.1016/j.ajog.2024.01.003] [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: 09/08/2023] [Revised: 12/22/2023] [Accepted: 01/01/2024] [Indexed: 01/15/2024]
Abstract
BACKGROUND The investigation of the fetal umbilical-portal venous system is based on the premise that congenital anomalies of this system may be related to adverse perinatal outcomes. Several small retrospective studies have reported an association between umbilical-portal-systemic venous shunts and intrauterine growth restriction. However, the prevalence of portosystemic shunts in the fetal growth restricted population is yet to be determined. OBJECTIVE The aims of this study were (1) to determine the prevalence of fetal umbilical-portal-systemic venous shunts in pregnancies complicated by intrauterine growth restriction and (2) to compare the perinatal and neonatal outcomes of pregnancies with intrauterine growth restriction with and without umbilical-portal-systemic venous shunts. STUDY DESIGN This was a prospective, cross-sectional study of pregnancies diagnosed with intrauterine growth restriction, as defined by the Society for Maternal-Fetal Medicine intrauterine growth restriction guidelines. All participants underwent a detailed anomaly scan, supplemented with a targeted scan of the fetal portal system. Venous shunts were diagnosed using color Doppler mode. The perinatal outcomes of pregnancies with intrauterine growth restriction with and without umbilical-portal-systemic venous shunts were compared. RESULTS A total of 150 cases with intrauterine growth restriction were recruited. The prevalence of umbilical-portal-systemic venous shunts in our cohort was 9.3% (n=14). When compared with the control group (intrauterine growth restriction without umbilical-portal-systemic venous shunts, n=136), the study group had a significantly lower mean gestational age at the time of intrauterine growth restriction diagnosis (29.7±5.6 vs 32.47±4.6 weeks of gestation; P=.036) and an earlier gestational age at delivery (33.50±6.0 vs 36.13±2.8; P=.005). The study group had a higher rate of fetal death (21.4% vs 0.7%; P<.001) and, accordingly, a lower rate of live births (71.4% vs 95.6%; P=.001). Additional associated fetal vascular anomalies were significantly more prevalent in the study group than in the control group (35.7% vs 4.4%; P≤.001). The rate of other associated anomalies was similar. The study group had a significantly lower rate of abnormal uterine artery Doppler indices (0% vs 40.4%; P=.011) and a higher rate of abnormal ductus venosus Doppler indices (64.3% vs 23%; P=.001). There were no cases of hypertensive disorders of pregnancy in the study group, whereas the control group had an incidence of 12.5% (P=.16). Other perinatal and neonatal outcomes were comparable. CONCLUSION Umbilical-portal-systemic venous shunt is a relatively common finding among fetuses with growth restriction. When compared with pregnancies with intrauterine growth restriction with a normal portal system, these pregnancies complicated by intrauterine growth restriction and an umbilical-portal-systemic venous shunt are associated with a different Doppler flow pattern, an increased risk for fetal death, earlier presentation of intrauterine growth restriction, a lower gestational age at delivery, additional congenital vascular anomalies, and a lower rate of pregnancy-induced hypertensive disorders. Meticulous sonographic evaluation of the portal system should be considered in the prenatal workup of intrauterine growth restriction, as umbilical-portal-systemic venous shunts may affect perinatal outcomes.
Collapse
Affiliation(s)
- Shelly Czeiger
- Obstetrics and Gynecology Ultrasound Unit, Department of Obstetrics and Gynecology, Sheba Medical Center, Tel-Hashomer, Israel; Department of Obstetrics and Gynecology, Mayanei HaYeshuha Medical Center, Bnei-Brak, Israel.
| | - Tal Weissbach
- Obstetrics and Gynecology Ultrasound Unit, Department of Obstetrics and Gynecology, Sheba Medical Center, Tel-Hashomer, Israel; Department of Obstetrics and Gynecology, Sheba Medical Center, Tel Hashomer, Israel
| | - Keren Zloto
- Department of Obstetrics and Gynecology, Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ariella Wiener
- Department of Obstetrics and Gynecology, Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Omer Nir
- Department of Obstetrics and Gynecology, Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Abeer Massarwa
- Obstetrics and Gynecology Ultrasound Unit, Department of Obstetrics and Gynecology, Sheba Medical Center, Tel-Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Boaz Weisz
- Obstetrics and Gynecology Ultrasound Unit, Department of Obstetrics and Gynecology, Sheba Medical Center, Tel-Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Fishel Bartal
- Department of Obstetrics and Gynecology, Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rakefet Yoeli Ulman
- Department of Obstetrics and Gynecology, Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yossi Bart
- Department of Obstetrics and Gynecology, Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Reuven Achiron
- Obstetrics and Gynecology Ultrasound Unit, Department of Obstetrics and Gynecology, Sheba Medical Center, Tel-Hashomer, Israel
| | - Zvi Kivilevitch
- Obstetrics and Gynecology Ultrasound Unit, Department of Obstetrics and Gynecology, Sheba Medical Center, Tel-Hashomer, Israel
| | - Shali Mazaki-Tovi
- Department of Obstetrics and Gynecology, Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eran Kassif
- Obstetrics and Gynecology Ultrasound Unit, Department of Obstetrics and Gynecology, Sheba Medical Center, Tel-Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
39
|
Ramirez Zegarra R, Ghi T, Lees C. Does the use of angiogenic biomarkers for the management of preeclampsia and fetal growth restriction improve outcomes?: Challenging the current status quo. Eur J Obstet Gynecol Reprod Biol 2024; 300:268-277. [PMID: 39053087 DOI: 10.1016/j.ejogrb.2024.07.042] [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: 06/30/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
Monitoring and timing of delivery in preterm preeclampsia and fetal growth restriction is one of the biggest challenges in Obstetrics. Finding the optimal time of delivery of these fetuses usually involves a trade-off between the severity of the disease and prematurity. So far, most clinical guidelines recommend the use of a combination between clinical, laboratory and ultrasound markers to guide the time of delivery. Angiogenic biomarkers, especially placental growth factor (PlGF) and soluble fms-like tyrosine kinase-1 (sFlt-1), have gained significant attention in recent years for their potential role in the prediction and diagnosis of placenta-related disorders including preeclampsia and fetal growth restriction. Another potential clinical application of the angiogenic biomarkers is for the differential diagnosis of patients with chronic kidney disease, as this condition shares similar clinical features with preeclampsia. Consequently, angiogenic biomarkers have been advocated as tools for monitoring and deciding the optimal time of the delivery of fetuses affected by placental dysfunction. In this clinical opinion, we critically review the available literature on PlGF and sFlt-1 for the surveillance and time of the delivery in fetuses affected by preterm preeclampsia and fetal growth restriction. Moreover, we explore the use of angiogenic biomarkers for the differentiation between chronic kidney disease and superimposed preeclampsia.
Collapse
Affiliation(s)
- Ruben Ramirez Zegarra
- Department of Medicine and Surgery, Obstetrics and Gynecology Unit, University of Parma, Parma, Italy
| | - Tullio Ghi
- Department of Medicine and Surgery, Obstetrics and Gynecology Unit, University of Parma, Parma, Italy
| | - Christoph Lees
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom; Centre for Fetal Care, Queen Charlotte's and Chelsea Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom; Department of Development and Regeneration, Katholieke Universiteit Leuven, Leuven, Belgium
| |
Collapse
|
40
|
Chatzakis C, Papavasiliou D, Mansukhani T, Nicolaides KH, Charakida M. Maternal vascular-placental axis in the third trimester in women with gestational diabetes mellitus, hypertensive disorders, and unaffected pregnancies. Am J Obstet Gynecol 2024:S0002-9378(24)00900-1. [PMID: 39218286 DOI: 10.1016/j.ajog.2024.08.045] [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: 04/13/2024] [Revised: 08/10/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Hypertensive disorders of pregnancy and gestational diabetes mellitus are characterized by vascular dysfunction and are associated with long term cardiovascular risks. OBJECTIVE This study aimed to compare different markers of maternal vascular function in women with gestational diabetes mellitus, preeclampsia, or gestational hypertension and in women whose pregnancies were unaffected by these complications and to assess the association between maternal vascular function and markers of placental perfusion and maternal vascular-placental axis in 4 groups of women. STUDY DESIGN This was a prospective observational study of women who had routine hospital visits at 35 0/7 to 36 6/7 weeks of gestation at King's College Hospital, London, United Kingdom. The routine hospital visit included recording of maternal demographic characteristics and medical history, ultrasound examination for fetal anatomy and growth, Doppler studies of the uterine arteries and ophthalmic arteries, carotid-femoral pulse wave velocity measurements, estimation of the augmentation index and total peripheral resistance, and measurements of serum placental growth factor and soluble fms-like tyrosine kinase 1. Linear regression analysis was performed for the outcomes of uterine artery pulsatility index multiple of the median, placental growth factor multiple of the median, and soluble fms-like tyrosine kinase 1 multiple of the median. The ophthalmic artery peak systolic velocity ratio, pulse wave velocity, augmentation index, and total peripheral vascular resistance were assessed as potential predictors. This analysis was performed on all women and separately in the different groups. RESULTS The study population of 6502 women included 614 (9.4%) with gestational diabetes mellitus, 140 (2.1%) who subsequently developed preeclampsia, and 129 (2.0%) who developed gestational hypertension. Women with gestational diabetes mellitus had increased pulse wave velocity compared with those with pregnancies unaffected by gestational diabetes mellitus, preeclampsia, or gestational hypertension. Women with preeclampsia or gestational hypertension had lower placental growth factor multiple of the median and higher uterine artery pulsatility index multiple of the median, soluble fms-like tyrosine kinase 1 multiple of the median, augmentation index, pulse wave velocity, total peripheral resistance, and ophthalmic artery peak systolic velocity ratio than those with unaffected pregnancies. In women with unaffected pregnancies, the ophthalmic artery peak systolic velocity ratio was predictive of the uterine artery pulsatility index multiple of the median, and ophthalmic artery peak systolic velocity ratio, augmentation index, total peripheral resistance, and pulse wave velocity were predictive of the placental growth factor multiple of the median and the soluble fms-like tyrosine kinase 1 multiple of the median. In women with gestational diabetes mellitus, the ophthalmic artery peak systolic velocity ratio was predictive of the uterine artery pulsatility index multiple of the median; the ophthalmic artery peak systolic velocity ratio, total peripheral resistance, and pulse wave velocity were predictive of the placental growth factor multiple of the median; and total peripheral resistance was predictive of the soluble fms-like tyrosine kinase 1 multiple of the median. In women with preeclampsia, the ophthalmic artery peak systolic velocity ratio was predictive of the uterine artery pulsatility index multiple of the median, placental growth factor multiple of the median, and soluble fms-like tyrosine kinase 1 multiple of the median. In women unaffected by gestational diabetes mellitus, preeclampsia, or gestational hypertension, the ophthalmic artery peak systolic velocity ratio was predictive of the uterine artery pulsatility index multiple of the median, and the augmentation index, total peripheral resistance, pulse wave velocity, and the ophthalmic artery peak systolic velocity ratio were predictive of the placental growth factor multiple of the median and the soluble fms-like tyrosine kinase 1 multiple of the median. CONCLUSION In the third trimester of pregnancy, women with preeclampsia, gestational hypertension, and gestational diabetes mellitus present with increased arterial stiffness. In addition, women diagnosed with hypertensive complications showed increased peripheral vascular resistance. The ophthalmic artery peak systolic velocity ratio provided predictive information for placental perfusion and function in all pregnant women, whereas vascular indices were more informative for placental function in women with unaffected pregnancies and those with gestational diabetes mellitus than in those with preeclampsia or gestational hypertension. Our data suggest that vascular assessment in women during pregnancy not only may provide information about maternal vascular health but also can be used to provide information about individual risk factors for placental insufficiency. The selection of the vascular index will have to be tailored according to the maternal profile and pregnancy complication.
Collapse
Affiliation(s)
- Christos Chatzakis
- Second Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - Dimitra Papavasiliou
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom
| | - Tanvi Mansukhani
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom
| | - Kypros H Nicolaides
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom.
| | - Marietta Charakida
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom; School of Biomedical Engineering and Imaging Sciences, King's College Hospital, London, United Kingdom
| |
Collapse
|
41
|
Naidoo SJ, Naicker T. The Enigmatic Interplay of Interleukin-10 in the Synergy of HIV Infection Comorbid with Preeclampsia. Int J Mol Sci 2024; 25:9434. [PMID: 39273381 PMCID: PMC11395227 DOI: 10.3390/ijms25179434] [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: 07/23/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Cytokines coordinate the intricate choreography of the immune system, directing cellular activities that mediate inflammation, pathogen defense, pathology and tissue repair. Within this spectrum, the anti-inflammatory prowess of interleukin-10 (IL-10) predominates in immune homeostasis. In normal pregnancy, the dynamic shift of IL-10 across trimesters maintains maternal immune tolerance ensuring fetal development and pregnancy success. Unravelling the dysregulation of IL-10 in pregnancy complications is vital, particularly in the heightened inflammatory condition of preeclampsia. Of note, a reduction in IL-10 levels contributes to endothelial dysfunction. In human immunodeficiency virus (HIV) infection, a complex interplay of IL-10 occurs, displaying a paradoxical paradigm of being immune-protective yet aiding viral persistence. Genetic variations in the IL-10 gene further modulate susceptibility to HIV infection and preeclampsia, albeit with nuanced effects across populations. This review outlines the conceptual framework underlying the role of IL-10 in the duality of normal pregnancy and preeclampsia together with HIV infection, thus highlighting its regulatory mechanisms and genetic influences. Synthesizing these findings in immune modulation presents avenues for therapeutic interventions in pregnancy complications comorbid with HIV infection.
Collapse
Affiliation(s)
| | - Thajasvarie Naicker
- Department of Optics and Imaging, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
| |
Collapse
|
42
|
Di Tonto A, Nogue L, Valentini B, Bennasar M, Melito C, Sorrentino S, Gómez O, Corno E, Baffa MT, Ghi T, Crispi F, Dall'Asta A. Reproducibility Analysis of Two Speckle Tracking Software for the Antenatal Semiautomated Assessment of the Fetal Cardiac Function. Fetal Diagn Ther 2024; 52:8-18. [PMID: 39159614 DOI: 10.1159/000541007] [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: 01/05/2024] [Accepted: 08/14/2024] [Indexed: 08/21/2024]
Abstract
INTRODUCTION Speckle tracking echocardiography is a non-Doppler modality allowing the semiautomated evaluation of the fetal cardiac function by tracking the speckles of the endocardial borders. Little evidence is available on the evaluation and comparison of different software for the functional assessment of the fetal heart by means of speckle tracking echocardiography. The aim of this study was to evaluate the reproducibility and agreement of two different proprietary speckle tracking software for the prenatal semiautomated assessment of the fetal cardiac function. METHODS The prospective study including non-anomalous fetuses was referred for different indications at two tertiary academic units in Italy (University of Parma) and Spain (University of Barcelona). Two-dimensional clips of the four-chamber view of the fetal heart were acquired by two dedicated operators using high-end ultrasound machines with a frame rate higher than 60 Hz. The stored clips were pseudo-anonymized and shared between the collaborating units. Functional echocardiographic analyses were independently performed using the two proprietary software (TomTec GmbH and FetalHQ®) by the same operators. Inter-software reproducibility of the endocardial global longitudinal strain (EndoGLS) and fractional area change (FAC) of the left (LV) and the right ventricles (RV) and ejection fraction (EF) of the LV were evaluated by the intraclass correlation coefficient (ICC). RESULTS Forty-eight fetuses were included at a median of 31+2 (21+6-40+3) gestational weeks. Moderate reproducibility was found for the functional parameters of the LV: EndoGLS (Pearson's correlation 0.456, p < 0.01; ICC 0.446, 95% CI: 0.189-0.647, p < 0.01); EF (Pearson's correlation 0.435, p < 0.01; ICC 0.419, 95% CI: 0.156-0.627, p < 0.01); FAC (Person's correlation 0.484, p < 0.01; ICC 0.475, 95% CI: 0.223-0.667, p < 0.01). On the contrary, RV functional parameters showed poor reproducibility between the two software: EndoGLS (Pearson's correlation 0.383, p = 0.01; ICC 0.377, 95% CI: 0.107-0.596, p < 0.01) and FAC (ICC 0.284, 95% CI: 0.003-0.524, p = 0.02). CONCLUSION Our results demonstrate a moderate reproducibility of the speckle tracking analysis of the LV using TomTec GmbH and FetalHQ®, with poor reproducibility for RV analysis.
Collapse
Affiliation(s)
- Andrea Di Tonto
- Department of Medicine and Surgery, Unit of Surgical Sciences, Obstetrics and Gynecology, University of Parma, Parma, Italy,
| | - Laura Nogue
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, CIBERER, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Beatrice Valentini
- Department of Medicine and Surgery, Unit of Surgical Sciences, Obstetrics and Gynecology, University of Parma, Parma, Italy
| | - Mar Bennasar
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, CIBERER, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Chiara Melito
- Department of Medicine and Surgery, Unit of Surgical Sciences, Obstetrics and Gynecology, University of Parma, Parma, Italy
| | - Sara Sorrentino
- Department of Medicine and Surgery, Unit of Surgical Sciences, Obstetrics and Gynecology, University of Parma, Parma, Italy
| | - Olga Gómez
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, CIBERER, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Enrico Corno
- Department of Medicine and Surgery, Unit of Surgical Sciences, Obstetrics and Gynecology, University of Parma, Parma, Italy
| | - Maria Teresa Baffa
- Department of Medicine and Surgery, Unit of Surgical Sciences, Obstetrics and Gynecology, University of Parma, Parma, Italy
| | - Tullio Ghi
- Department of Medicine and Surgery, Unit of Surgical Sciences, Obstetrics and Gynecology, University of Parma, Parma, Italy
| | - Fàtima Crispi
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, CIBERER, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Andrea Dall'Asta
- Department of Medicine and Surgery, Unit of Surgical Sciences, Obstetrics and Gynecology, University of Parma, Parma, Italy
| |
Collapse
|
43
|
Uçkan K, Özgökçe Ç, Başkiran Y, Eyisoy ÖG, Çeleğen İ, Akbay Hİ. The role of ultrasound and mitofusin-2 levels to predict pregnancy outcomes in patients with severe preeclampsia: a case-control study. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20240152. [PMID: 39166673 PMCID: PMC11329264 DOI: 10.1590/1806-9282.20240152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 08/23/2024]
Abstract
OBJECTIVE The aim of this study was to evaluate mitofusin-2 levels and fetal Doppler ultrasonography effects in patients with severe preeclampsia. METHODS This single-center case-control study was conducted in the gynecology service of the university hospital in Van. A total of 90 pregnant women aged 18-40 years were included in the study. Of these, 30 are normal, 30 have mild preeclampsia, and 30 are pregnant with severe preeclampsia. In this study, especially in severe preeclampsia patients, serum mitofusin-2 levels and important fetal Doppler flows such as uterine arterial pressure, umbilical arterial pressure, and 1st and 5th minute Apgar scores, birth weight, and the relationship between postnatal outcomes such as week of birth and the number of patients in the neonatal intensive care unit were investigated. RESULTS There was a significant difference between the three groups in terms of mitofusin-2 levels, which was the highest in the group (p<0.05). Maternal serum mitofusin-2 levels were positively correlated with uterine arterial pressure (r=0.543, p=0.007), umbilical arterial pressure (r=0.238, p=0.008), diastolic blood pressure, and systolic blood pressure (p<0.001). Receiver operating characteristic curve of mitofusin-2 in predicting preeclampsia is as follows: optimal cutoff 1.6 ng/mL; area under the curve: 0.861; 95%CI: 0.786-0.917; sensitivity: 83.9%; and specificity: 70.0%, (p≤0.001). A one-unit increase in mitofusin-2 resulted in a statistically significant 4.21-fold increase in preeclampsia risk. CONCLUSION This study recommends the use of mitofusin-2 together with fetal Doppler ultrasound findings as a reliable indicator of preeclampsia severity.
Collapse
Affiliation(s)
- Kazım Uçkan
- Yuzuncu Yil University, Faculty of Medicine, Gynecology and Obstetrics Clinic - Van, Turkey
| | - Çağdaş Özgökçe
- Zeynep Kamil Women and Children Research Hospital - İstanbul, Turkey
| | - Yusuf Başkiran
- Yuzuncu Yil University, Faculty of Medicine, Gynecology and Obstetrics Clinic - Van, Turkey
| | - Ömer Gökhan Eyisoy
- Yuzuncu Yil University, Faculty of Medicine, Gynecology and Obstetrics Clinic - Van, Turkey
| | - İzzet Çeleğen
- Yuzuncu Yil University, Faculty of Medicine, Department of Public Health - Van, Turkey
| | - Halil İbrahim Akbay
- Yuzuncu Yil University, Faculty of Medicine, Gynecology and Obstetrics Clinic - Van, Turkey
| |
Collapse
|
44
|
Chen X, Lan L, Wu H, Zeng M, Zheng Z, Zhong Q, Lai F, Hu Y. Chromosomal Microarray Analysis in Fetuses with Ultrasound Abnormalities. Int J Gen Med 2024; 17:3531-3540. [PMID: 39161407 PMCID: PMC11332413 DOI: 10.2147/ijgm.s472906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/09/2024] [Indexed: 08/21/2024] Open
Abstract
Objective To explore and evaluate the value of chromosomal microarray analysis (CMA) in prenatal diagnosis of fetuses with ultrasound abnormalities. Methods A retrospective analysis was performed on 370 fetuses with ultrasound abnormalities received invasive prenatal diagnosis at Meizhou People's Hospital from October 2022 to December 2023. Fetal specimens were analyzed by CMA, and the detection rates of aneuploidy and pathogenic (P)/likely pathogenic (LP) copy number variations (CNVs) in ultrasound structural abnormalities (malformations of fetal anatomy) and non-structural abnormalities (abnormalities of fetal nonanatomical structure) were analyzed. Results There were 114 (30.8%) cases with isolated ultrasound structural abnormalities, 226 (61.1%) cases with isolated non-structural abnormalities (182 isolated ultrasound soft markers abnormalities, 30 isolated fetal growth restriction (FGR), and 8 isolated abnormalities of amniotic fluid volume), and 30 (8.1%) cases with both structural and non-structural abnormalities. The overall detection rate of aneuploidy and P/LP CNVs in isolated ultrasonic structural abnormalities was 5.3%, among which cardiovascular system abnormalities were the highest. In addition, the largest number of fetuses with non-structural abnormalities was nuchal translucency (NT) thickening (n = 81), followed by ventriculomegaly (n = 29), and nasal bone dysplasia (n = 24). The detection rate of chromosomal abnormalities of fetuses with abnormal ultrasound soft markers was 9.9%, and the detection rate in single abnormal ultrasound soft marker, and multiple ultrasound soft markers abnormalities was 9.7% (16/165) and 11.8% (2/17), respectively. Moreover, the detection rate of chromosomal abnormalities of fetuses with FGR and structural abnormalities combined with non-structural abnormalities was 6.7% (2/30), and 13.3% (4/30), respectively. Conclusion The incidence of chromosomal abnormalities (aneuploidy and P/LP CNVs) varies among different fetal ultrasound abnormalities.
Collapse
Affiliation(s)
- Xiaoqin Chen
- Department of Prenatal Diagnostic Center, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
- Department of Obstetrics, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| | - Liubing Lan
- Department of Prenatal Diagnostic Center, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
- Department of Obstetrics, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| | - Heming Wu
- Department of Prenatal Diagnostic Center, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| | - Mei Zeng
- Department of Prenatal Diagnostic Center, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
- Department of Obstetrics, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| | - Zhiyuan Zheng
- Department of Prenatal Diagnostic Center, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| | - Qiuping Zhong
- Department of Obstetrics, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| | - Fengdan Lai
- Department of Obstetrics, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| | - Yonghe Hu
- Department of Obstetrics, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, People’s Republic of China
| |
Collapse
|
45
|
Wei XH, Liao LY, Yin YX, Xu Q, Xie SS, Liu M, Gao LB, Chen HQ, Zhou R. Overexpression of long noncoding RNA DUXAP8 inhibits ER-phagy through activating AKT/mTOR signaling and contributes to preeclampsia. Cell Mol Life Sci 2024; 81:336. [PMID: 39120751 PMCID: PMC11335266 DOI: 10.1007/s00018-024-05385-y] [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: 04/05/2024] [Revised: 07/09/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
Preeclampsia (PE) is a life-threatening pregnancy-specific complication with controversial mechanisms and no effective treatment except delivery is available. Currently, increasing researchers suggested that PE shares pathophysiologic features with protein misfolding/aggregation disorders, such as Alzheimer disease (AD). Evidences have proposed defective autophagy as a potential source of protein aggregation in PE. Endoplasmic reticulum-selective autophagy (ER-phagy) plays a critical role in clearing misfolded proteins and maintaining ER homeostasis. However, its roles in the molecular pathology of PE remain unclear. We found that lncRNA DUXAP8 was upregulated in preeclamptic placentae and significantly correlated with clinical indicators. DUXAP8 specifically binds to PCBP2 and inhibits its ubiquitination-mediated degradation, and decreased levels of PCBP2 reversed the activation effect of DUXAP8 overexpression on AKT/mTOR signaling pathway. Function experiments showed that DUXAP8 overexpression inhibited trophoblastic proliferation, migration, and invasion of HTR-8/SVneo and JAR cells. Moreover, pathological accumulation of swollen and lytic ER (endoplasmic reticulum) was observed in DUXAP8-overexpressed HTR8/SVneo cells and PE placental villus trophoblast cells, which suggesting that ER clearance ability is impaired. Further studies found that DUXAP8 overexpression impaired ER-phagy and caused protein aggregation medicated by reduced FAM134B and LC3II expression (key proteins involved in ER-phagy) via activating AKT/mTOR signaling pathway. The increased level of FAM134B significantly reversed the inhibitory effect of DUXAP8 overexpression on the proliferation, migration, and invasion of trophoblasts. In vivo, DUXAP8 overexpression through tail vein injection of adenovirus induced PE-like phenotypes in pregnant rats accompanied with activated AKT/mTOR signaling, decreased expression of FAM134B and LC3-II proteins and increased protein aggregation in placental tissues. Our study reveals the important role of lncRNA DUXAP8 in regulating trophoblast biological behaviors through FAM134B-mediated ER-phagy, providing a new theoretical basis for understanding the pathogenesis of PE.
Collapse
Affiliation(s)
- Xiao-Hong Wei
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) , Ministry of Education, West China Second University Hospital, Sichuan University, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Ling-Yun Liao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) , Ministry of Education, West China Second University Hospital, Sichuan University, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Yang-Xue Yin
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) , Ministry of Education, West China Second University Hospital, Sichuan University, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Qin Xu
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) , Ministry of Education, West China Second University Hospital, Sichuan University, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Shuang-Shuang Xie
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) , Ministry of Education, West China Second University Hospital, Sichuan University, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Min Liu
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) , Ministry of Education, West China Second University Hospital, Sichuan University, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Lin-Bo Gao
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Hong-Qin Chen
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) , Ministry of Education, West China Second University Hospital, Sichuan University, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Rong Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) , Ministry of Education, West China Second University Hospital, Sichuan University, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, P.R. China.
| |
Collapse
|
46
|
Rottenstreich A. Placenta-Mediated Conditions: Past, Present, and Future Perspectives. J Clin Med 2024; 13:4631. [PMID: 39200773 PMCID: PMC11355048 DOI: 10.3390/jcm13164631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Pregnancy is a highly regulated biological condition in which a successful outcome is heavily dependent on maintaining a delicate balance through maternal-fetal dialog at various levels [...].
Collapse
Affiliation(s)
- Amihai Rottenstreich
- Laboratory of Blood and Vascular Biology, Rockefeller University, New York, NY 10065, USA; ; Tel.: +212-327-7494; Fax: 212-327-7493
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Zucker School of Medicine at Hofstra/Northwell, New York, NY, USA
| |
Collapse
|
47
|
Giorgione V, Ramnarine S, Malik A, Bhide A. The value of angiogenetic biomarkers in the detection of early onset fetal growth restriction. Eur J Obstet Gynecol Reprod Biol 2024; 299:91-95. [PMID: 38850897 DOI: 10.1016/j.ejogrb.2024.05.036] [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: 01/09/2024] [Revised: 05/01/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
OBJECTIVE The identification of fetal growth restriction (FGR) due to uteroplacental insufficiency is important to improve perinatal outcomes. To distinguish FGR from small for gestational age (SGA), FGR consensus definition is currently based on biometry and/or additional biophysical parameters. This study aims to verify if this definition might be modified by including circulating angiogenic factors. STUDY DESIGN This historical cohort study included singleton pregnancies with SGA fetuses after 20 weeks. All patients underwent detailed ultrasound and measurements of soluble fms-like tyrosine kinase 1 (sFlt-1) and placental growth factor (PlGF) at first assessment. ISUOG criteria for FGR were applied. Total PlGF was calculated using free PlGF, sFlt-1 and a receptor pharmacology model, and multiple of the median (MoM) values for sFlt-1, free PlGF, total PlGF and sFlt-1/PlGF ratio were calculated to adjust for gestational age. RESULTS 72 pregnancies with SGA were first evaluated at median (IQR) of 28+5 (26+2 -31+3) weeks' gestation, and 51 fetuses (70.8 %) satisfied the FGR consensus definition. Pregnancies with FGR showed significantly lower levels of free and total PlGF MoM (0.12, 95 % IQR: 0.07-0.36 vs 0.32, 95 % IQR: 0.20-0.53, p = 0.008) and 0.26, 95 % CI: 0.16-0.55 vs 0.43, 95 % IQR: 0.23-0.53, p = 0.028) respectively; and higher sFlt-1 MoM (4.62, 95 % IQR: 1.80-7.30 vs 1.74, 95 % IQR:1.11-3.61, p = 0.014) than pregnancies not classified as FGR. Free and total PlGF MoM correlated significantly with gestational age at delivery (r = 0.776, p < 0.001 and r = 0.707, p < 0.001, respectively). sFlt-1 MoM and sFlt-1/PlGF ratio MoM also correlated with gestational age at delivery (r = -0.681, p < 0.001 and r = -0.823, p < 0.001). Six cases identified as FGR at first ultrasound were not confirmed at birth showing significantly higher levels of free PlGF MoM (0.77, 95 % IQR: 0.27-3.07 vs 0.17, 95 % IQR: 0.08-0.43, p = 0.022). CONCLUSION These findings show that total as well as free PlGF levels are lower in pregnancies affected with placental growth restriction. Angiogenic biomarkers might improve the differentiation between placental growth restriction and constitutional smallness. Further studies are needed to determine how to integrate them into the current definitions of FGR.
Collapse
Affiliation(s)
- Veronica Giorgione
- Maternity Department, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Stephan Ramnarine
- Maternity Department, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Amna Malik
- Maternity Department, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Amarnath Bhide
- Maternity Department, St George's University Hospitals NHS Foundation Trust, London, UK; Vascular Biology Research Center, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK.
| |
Collapse
|
48
|
Silver RM, Reddy U. Stillbirth: we can do better. Am J Obstet Gynecol 2024; 231:152-165. [PMID: 38789073 DOI: 10.1016/j.ajog.2024.05.042] [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: 12/27/2023] [Revised: 04/23/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024]
Abstract
Stillbirth is far too common, occurring in millions of pregnancies per year globally. The rate of stillbirth (defined as death of a fetus prior to birth at 20 weeks' gestation or more) in the United States is 5.73 per 1000. This is approximately 1 in 175 pregnancies accounting for about 21,000 stillbirths per year. Although rates are much higher in low-income countries, the stillbirth rate in the United States is much higher than most high resource countries. Moreover, there are substantial disparities in stillbirth, with rates twice as high for non-Hispanic Black and Native Hawaiian or Other Pacific Islanders compared to non-Hispanic Whites. There is considerable opportunity for reduction in stillbirths, even in high resource countries such as the United States. In this article, we review the epidemiology, risk factors, causes, evaluation, medical and emotional management, and prevention of stillbirth. We focus on novel data regarding genetic etiologies, placental assessment, risk stratification, and prevention.
Collapse
Affiliation(s)
- Robert M Silver
- Departments of Obstetrics and Gynecology, Divisions of Maternal Fetal Medicine, University of Utah, Salt Lake City, UT.
| | - Uma Reddy
- Departments of Obstetrics and Gynecology, Divisions of Maternal Fetal Medicine, Columbia University, New York, NY
| |
Collapse
|
49
|
Moronge D, Ayulo V, Elgazzaz M, Mellott E, Ogbi S, Faulkner JL. Both endothelial mineralocorticoid receptor expression and hyperleptinemia are required for clinical characteristics of placental ischemia in mice. Am J Physiol Heart Circ Physiol 2024; 327:H118-H130. [PMID: 38758130 PMCID: PMC11380964 DOI: 10.1152/ajpheart.00188.2024] [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: 03/26/2024] [Revised: 04/23/2024] [Accepted: 05/09/2024] [Indexed: 05/18/2024]
Abstract
One of the initiating events in preeclampsia (PE) is placental ischemia. Rodent models of placental ischemia do not present with vascular endothelial dysfunction, a hallmark of PE. We previously demonstrated a role for leptin in endothelial dysfunction in pregnancy in the absence of placental ischemia. We hypothesized that placental ischemia requires hyperleptinemia and endothelial mineralocorticoid receptor (ECMR) expression to induce PE-associated endothelial dysfunction in pregnant mice. We induced placental ischemia via the reduced uterine perfusion pressure (RUPP) procedure in pregnant ECMR-intact (ECMR+/+) and ECMR deletion (ECMR-/-) mice at gestational day (GD) 13. ECMR+/+ RUPP pregnant mice also received concurrent leptin infusion via miniosmotic pump (0.9 mg/kg/day). RUPP increased blood pressure via radiotelemetry and decreased fetal growth in ECMR+/+ pregnant mice. Both increases in blood pressure and reduced fetal growth were abolished in RUPP ECMR-/- mice. Placental ischemia did not decrease endothelial-dependent relaxation to acetylcholine (ACh) but increased phenylephrine (Phe) contraction in mesenteric arteries of pregnant mice, which was ablated by ECMR deletion. Addition of leptin to RUPP mice significantly reduced ACh relaxation in ECMR+/+ pregnant mice, accompanied by an increase in soluble FMS-like tyrosine kinase-1 (sFlt-1)/placental growth factor (PLGF) ratio. In conclusion, our data indicate that high leptin levels drive endothelial dysfunction in PE and that ECMR is required for clinical characteristics of hypertension and fetal growth restriction in placental ischemia PE. Collectively, we show that both ECMR and leptin play a role to mediate PE.NEW & NOTEWORTHY Leptin is a key feature of preeclampsia that initiates vascular endothelial dysfunction in preeclampsia characterized by placental ischemia. Endothelial mineralocorticoid receptor (ECMR) deletion in placental ischemia protects pregnant mice from elevations in blood pressure and fetal growth restriction in pregnancy. Increases in leptin production mediate the key pathological feature of endothelial dysfunction in preeclampsia in rodents. ECMR activation contributes to the increase in blood pressure and fetal growth restriction in preeclampsia.
Collapse
Affiliation(s)
- Desmond Moronge
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Victor Ayulo
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Department of Pediatrics, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Mona Elgazzaz
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Elisabeth Mellott
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Safia Ogbi
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Jessica L Faulkner
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Department of Obstetrics and Gynecology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| |
Collapse
|
50
|
Nagao T, Hasegawa A, Samura O, Okamoto A. Utilizing Repetitive Serial Placental Ultrasounds for Diagnosing a Massive Subchorionic Thrombohematoma: Insights Into Soluble FMS-Like Tyrosine Kinase-1/Placental Growth Factor (sFlt-1/PlGF) Trends. Cureus 2024; 16:e63927. [PMID: 39105034 PMCID: PMC11299127 DOI: 10.7759/cureus.63927] [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] [Accepted: 07/04/2024] [Indexed: 08/07/2024] Open
Abstract
The patient, a 34-year-old primigravida with no prior medical history, presented at 23 + 0 weeks with gestational hypertension and fetal growth restriction (FGR). Ultrasound examination showed a placental mass, and subsequent repeated ultrasound scans revealed changes in the mass' echogenicity, raising suspicion of a massive subchorionic thrombohematoma (MST). While the blood pressure was mildly elevated without proteinuria and organ dysfunctions, serum soluble fms-like tyrosine kinase-1/placental growth factor (sFlt-1/PlGF) ratios showed significantly elevated values. A cesarean section was performed at 29 + 2 weeks due to the nonreassuring fetal status. The female infant, with Apgar scores of 1/1 at one/five minutes and an umbilical artery pH of 7.16, remained unresponsive and died seven hours postdelivery. Pathology examination revealed a massive hematoma in the subchorionic space, measuring 22 mm thick, directly beneath the umbilical cord attachment. This case underscores the importance of repetitive placental ultrasound in MST diagnosis and suggests the potential utility of sFlt-1/PlGF ratios in predicting adverse outcomes.
Collapse
Affiliation(s)
- Takeshi Nagao
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, JPN
| | - Akihiro Hasegawa
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, JPN
| | - Osamu Samura
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, JPN
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, JPN
| |
Collapse
|