101
|
Khosla K, Jin Y, Espinoza J, Kent M, Gencay M, Kunz LH, Mueller A, Xiao Y, Frank Peacock W, Neath SX, Stuart JJ, Woelkers D, Harris JM, Rana S. Signs or symptoms of suspected preeclampsia - A retrospective national database study of prevalence, costs, and outcomes. Pregnancy Hypertens 2024; 36:101124. [PMID: 38608393 DOI: 10.1016/j.preghy.2024.101124] [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: 07/07/2023] [Revised: 04/06/2024] [Accepted: 04/07/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Most patients with signs or symptoms (s/s) of suspected preeclampsia are not diagnosed with preeclampsia. We sought to determine and compare the prevalence of s/s, pregnancy outcomes, and costs between patients with and without diagnosed preeclampsia. METHODS This retrospective cohort study analyzed a large insurance research database. Pregnancies with s/s of preeclampsia versus a confirmed preeclampsia diagnosis were identified using International Classification of Diseases codes. S/s include hypertension, proteinuria, headache, visual symptoms, edema, abdominal pain, and nausea/vomiting. Pregnancies were classed as 1) s/s of preeclampsia without a confirmed preeclampsia diagnosis (suspicion only), 2) s/s with a confirmed diagnosis (preeclampsia with suspicion), 3) diagnosed preeclampsia without s/s recorded (preeclampsia only), and 4) no s/s, nor preeclampsia diagnosis (control). RESULTS Of 1,324,424 pregnancies, 29.2 % had ≥1 documented s/s of suspected preeclampsia, and 14.2 % received a preeclampsia diagnosis. Hypertension and headache were the most common s/s, leading 20.2 % and 9.2 % pregnancies developed to preeclampsia diagnosis, respectively. Preeclampsia, with or without suspicion, had the highest rates of hypertension-related severe maternal morbidity (HR [95 % CI]: 3.0 [2.7, 3.2] and 3.6 [3.3, 4.0], respectively) versus controls. A similar trend was seen in neonatal outcomes such as preterm delivery and low birth weight. Cases in which preeclampsia was suspected but not confirmed had the highest average total maternal care costs ($6096 [95 % CI: 602, 6170] over control). CONCLUSION There is a high prevalence but poor selectivity of traditional s/s of preeclampsia, highlighting a clinical need for improved screening method and cost-effectiveness disease management.
Collapse
Affiliation(s)
- Kavia Khosla
- University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Yue Jin
- Roche Diagnostics, Indianapolis, IN, USA
| | - Jimmy Espinoza
- Department of Obstetrics and Gynecology, Division of Fetal Intervention, McGovern Medical School at the University of Texas Health Science Center Houston, and UT Physicians, The Fetal Center, Affiliated with Children's Memorial Hermann Hospital, TX, USA
| | - Matthew Kent
- Department of Epidemiology and Biostatistics, Genesis Research, Hoboken, NJ, USA
| | | | - Liza H Kunz
- Roche Diagnostics Systems, San Jose, CA, USA
| | - Ariel Mueller
- University of Chicago Pritzker School of Medicine, Chicago, IL, USA; Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yan Xiao
- Roche Diagnostics Systems, San Jose, CA, USA
| | - W Frank Peacock
- Department of Emergency Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Sean-Xavier Neath
- Department of Emergency Medicine, Gynecology and Reproductive Science, University of California, La Jolla, CA, USA
| | - Jennifer J Stuart
- Division of Women's Health, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Douglas Woelkers
- Department of Obstetrics, Gynecology and Reproductive Science, University of California, La Jolla, CA, USA
| | | | - Sarosh Rana
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
102
|
Li Y, Sang Y, Chang Y, Xu C, Lin Y, Zhang Y, Chiu PCN, Yeung WSB, Zhou H, Dong N, Xu L, Chen J, Zhao W, Liu L, Yu D, Zang X, Ye J, Yang J, Wu Q, Li D, Wu L, Du M. A Galectin-9-Driven CD11c high Decidual Macrophage Subset Suppresses Uterine Vascular Remodeling in Preeclampsia. Circulation 2024; 149:1670-1688. [PMID: 38314577 DOI: 10.1161/circulationaha.123.064391] [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: 02/13/2023] [Accepted: 01/09/2024] [Indexed: 02/06/2024]
Abstract
BACKGROUND Preeclampsia is a serious disease of pregnancy that lacks early diagnosis methods or effective treatment, except delivery. Dysregulated uterine immune cells and spiral arteries are implicated in preeclampsia, but the mechanistic link remains unclear. METHODS Single-cell RNA sequencing and spatial transcriptomics were used to identify immune cell subsets associated with preeclampsia. Cell-based studies and animal models including conditional knockout mice and a new preeclampsia mouse model induced by recombinant mouse galectin-9 were applied to validate the pathogenic role of a CD11chigh subpopulation of decidual macrophages (dMφ) and to determine its underlying regulatory mechanisms in preeclampsia. A retrospective preeclampsia cohort study was performed to determine the value of circulating galectin-9 in predicting preeclampsia. RESULTS We discovered a distinct CD11chigh dMφ subset that inhibits spiral artery remodeling in preeclampsia. The proinflammatory CD11chigh dMφ exhibits perivascular enrichment in the decidua from patients with preeclampsia. We also showed that trophoblast-derived galectin-9 activates CD11chigh dMφ by means of CD44 binding to suppress spiral artery remodeling. In 3 independent preeclampsia mouse models, placental and plasma galectin-9 levels were elevated. Galectin-9 administration in mice induces preeclampsia-like phenotypes with increased CD11chigh dMφ and defective spiral arteries, whereas galectin-9 blockade or macrophage-specific CD44 deletion prevents such phenotypes. In pregnant women, increased circulating galectin-9 levels in the first trimester and at 16 to 20 gestational weeks can predict subsequent preeclampsia onset. CONCLUSIONS These findings highlight a key role of a distinct perivascular inflammatory CD11chigh dMφ subpopulation in the pathogenesis of preeclampsia. CD11chigh dMφ activated by increased galectin-9 from trophoblasts suppresses uterine spiral artery remodeling, contributing to preeclampsia. Increased circulating galectin-9 may be a biomarker for preeclampsia prediction and intervention.
Collapse
Affiliation(s)
- Yanhong Li
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China (Y. Li, Y.S., C.X., Y. Lin, L.X., J.C., W.Z., L.L., D.L., M.D.)
- Department of Obstetrics, Longgang District Maternity and Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Clinical Institute of Shantou University Medical College), Shenzhen, Guangdong, China (Y. Li, Y. Lin, W.Z., J. Yang, M.D.)
- Department of Obstetrics and Gynecology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University Shanghai, China (Y. Li, M.D.)
| | - Yifei Sang
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China (Y. Li, Y.S., C.X., Y. Lin, L.X., J.C., W.Z., L.L., D.L., M.D.)
| | - Yunjian Chang
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (Y.C., Y.Z., H.Z., L.W.)
| | - Chunfang Xu
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China (Y. Li, Y.S., C.X., Y. Lin, L.X., J.C., W.Z., L.L., D.L., M.D.)
| | - Yikong Lin
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China (Y. Li, Y.S., C.X., Y. Lin, L.X., J.C., W.Z., L.L., D.L., M.D.)
| | - Yao Zhang
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (Y.C., Y.Z., H.Z., L.W.)
| | - Philip C N Chiu
- Department of Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, China (P.C.N.C., W.S.B.Y.)
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China (P.C.N.C., W.S.B.Y.)
| | - William S B Yeung
- Department of Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, China (P.C.N.C., W.S.B.Y.)
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China (P.C.N.C., W.S.B.Y.)
| | - Haisheng Zhou
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (Y.C., Y.Z., H.Z., L.W.)
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China (N.D., Q.W.)
| | - Ling Xu
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China (Y. Li, Y.S., C.X., Y. Lin, L.X., J.C., W.Z., L.L., D.L., M.D.)
| | - Jiajia Chen
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China (Y. Li, Y.S., C.X., Y. Lin, L.X., J.C., W.Z., L.L., D.L., M.D.)
| | - Weijie Zhao
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China (Y. Li, Y.S., C.X., Y. Lin, L.X., J.C., W.Z., L.L., D.L., M.D.)
- Department of Obstetrics, Longgang District Maternity and Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Clinical Institute of Shantou University Medical College), Shenzhen, Guangdong, China (Y. Li, Y. Lin, W.Z., J. Yang, M.D.)
| | - Lu Liu
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China (Y. Li, Y.S., C.X., Y. Lin, L.X., J.C., W.Z., L.L., D.L., M.D.)
| | - Di Yu
- The University of Queensland Diamantina Institute (D.Y.), Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre (D.Y.), Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY (X.Z.)
| | - Jiangfeng Ye
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research, Singapore City, Singapore (J. Ye)
| | - Jinying Yang
- Department of Obstetrics, Longgang District Maternity and Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Clinical Institute of Shantou University Medical College), Shenzhen, Guangdong, China (Y. Li, Y. Lin, W.Z., J. Yang, M.D.)
| | - Qingyu Wu
- Cyrus Tang Hematology Center, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China (N.D., Q.W.)
| | - Dajin Li
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China (Y. Li, Y.S., C.X., Y. Lin, L.X., J.C., W.Z., L.L., D.L., M.D.)
| | - Ligang Wu
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China (Y.C., Y.Z., H.Z., L.W.)
| | - Meirong Du
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China (Y. Li, Y.S., C.X., Y. Lin, L.X., J.C., W.Z., L.L., D.L., M.D.)
- Department of Obstetrics, Longgang District Maternity and Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Clinical Institute of Shantou University Medical College), Shenzhen, Guangdong, China (Y. Li, Y. Lin, W.Z., J. Yang, M.D.)
- Department of Obstetrics and Gynecology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University Shanghai, China (Y. Li, M.D.)
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau SAR, China (M.D.)
| |
Collapse
|
103
|
Zhu S, Ren J, Feng L, Jiang Y. Systemic Lupus Erythematosus and Pregnancy Complications and Outcomes: A Mendelian Randomization Study and Retrospective Validation. Int J Womens Health 2024; 16:891-902. [PMID: 38779383 PMCID: PMC11110830 DOI: 10.2147/ijwh.s461640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
Introduction Previous studies have shown that pregnant women with systemic lupus erythematosus (SLE) tend to have a higher risk of adverse pregnancy outcomes, but the potential causal role remained unclear. In this study, we aimed to investigate the causal relationship between SLE and some common pregnancy complications and outcomes using two-sample Mendelian randomization (MR). Methods The genetic tools were derived from genome-wide association studies of SLE and pregnancy complications and outcomes. MR analysis was performed using inverse variance weighting as primary method. Sensitivity analyses were performed to evaluate the robustness of the results. A retrospective analysis was conducted on 200 pregnant women with SLE and a control group of pregnant women delivering at Tongji Hospital. Results In the results, we found that genetic susceptibility to SLE was associated with a higher risk of gestational diabetes mellitus (OR = 1.028, 95% CI: 1.006-1.050), premature delivery (OR = 1.039, 95% CI: 1.013-1.066), polyhydramnios (OR = 1.075, 95% CI: 1.004-1.151) and premature rupture of membranes (OR = 1.030, 95% CI: 1.001-1.060). Some of the retrospective analysis results align with the findings from the MR analysis, indicating that pregnant women with SLE have a higher risk of developing gestational diabetes mellitus and preterm birth. Additionally, although MR analysis did not reveal a causal relationship between SLE and preeclampsia/eclampsia, retrospective analysis discovered that SLE pregnant women are more susceptible to developing preeclampsia/eclampsia (OR = 2.935, 95% CI: 1.118-7.620). Conclusion Our study findings suggest a potential causal relationship between SLE and increased risks of gestational diabetes and preterm delivery. Clinical data indicate that pregnant women with SLE are more prone to developing preeclampsia/eclampsia. Clinicians need to be vigilant about the occurrence of these conditions when managing pregnant women with SLE.
Collapse
Affiliation(s)
- Shenglan Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Junlin Ren
- Department of Computer Science, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
| | - Ling Feng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yi Jiang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| |
Collapse
|
104
|
Hoo R, Ruiz-Morales ER, Kelava I, Rawat M, Mazzeo CI, Tuck E, Sancho-Serra C, Chelaghma S, Predeus AV, Murray S, Fernandez-Antoran D, Waller RF, Álvarez-Errico D, Lee MCS, Vento-Tormo R. Acute response to pathogens in the early human placenta at single-cell resolution. Cell Syst 2024; 15:425-444.e9. [PMID: 38703772 DOI: 10.1016/j.cels.2024.04.002] [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: 05/19/2023] [Revised: 12/01/2023] [Accepted: 04/16/2024] [Indexed: 05/06/2024]
Abstract
The placenta is a selective maternal-fetal barrier that provides nourishment and protection from infections. However, certain pathogens can attach to and even cross the placenta, causing pregnancy complications with potential lifelong impacts on the child's health. Here, we profiled at the single-cell level the placental responses to three pathogens associated with intrauterine complications-Plasmodium falciparum, Listeria monocytogenes, and Toxoplasma gondii. We found that upon exposure to the pathogens, all placental lineages trigger inflammatory responses that may compromise placental function. Additionally, we characterized the responses of fetal macrophages known as Hofbauer cells (HBCs) to each pathogen and propose that they are the probable niche for T. gondii. Finally, we revealed how P. falciparum adapts to the placental microenvironment by modulating protein export into the host erythrocyte and nutrient uptake pathways. Altogether, we have defined the cellular networks and signaling pathways mediating acute placental inflammatory responses that could contribute to pregnancy complications.
Collapse
Affiliation(s)
- Regina Hoo
- Wellcome Sanger Institute, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | | | - Iva Kelava
- Wellcome Sanger Institute, Cambridge, UK
| | - Mukul Rawat
- Wellcome Sanger Institute, Cambridge, UK; Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, UK
| | | | | | | | - Sara Chelaghma
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | | - David Fernandez-Antoran
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK; Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ross F Waller
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Marcus C S Lee
- Wellcome Sanger Institute, Cambridge, UK; Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, UK.
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
105
|
Raja Xavier JP, Rianna C, Hellwich E, Nikolou I, Lankapalli AK, Brucker SY, Singh Y, Lang F, Schäffer TE, Salker MS. Excessive endometrial PlGF- Rac1 signalling underlies endometrial cell stiffness linked to pre-eclampsia. Commun Biol 2024; 7:530. [PMID: 38704457 PMCID: PMC11069541 DOI: 10.1038/s42003-024-06220-7] [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: 04/26/2023] [Accepted: 04/19/2024] [Indexed: 05/06/2024] Open
Abstract
Cell stiffness is regulated by dynamic interaction between ras-related C3 botulinum toxin substrate 1 (Rac1) and p21 protein-activated kinase 1 (PAK1) proteins, besides other biochemical and molecular regulators. In this study, we investigated how the Placental Growth Factor (PlGF) changes endometrial mechanics by modifying the actin cytoskeleton at the maternal interface. We explored the global effects of PlGF in endometrial stromal cells (EnSCs) using the concerted approach of proteomics, atomic force microscopy (AFM), and electrical impedance spectroscopy (EIS). Proteomic analysis shows PlGF upregulated RhoGTPases activating proteins and extracellular matrix organization-associated proteins in EnSCs. Rac1 and PAK1 transcript levels, activity, and actin polymerization were significantly increased with PlGF treatment. AFM further revealed an increase in cell stiffness with PlGF treatment. The additive effect of PlGF on actin polymerization was suppressed with siRNA-mediated inhibition of Rac1, PAK1, and WAVE2. Interestingly, the increase in cell stiffness by PlGF treatment was pharmacologically reversed with pravastatin, resulting in improved trophoblast cell invasion. Taken together, aberrant PlGF levels in the endometrium can contribute to an altered pre-pregnancy maternal microenvironment and offer a unifying explanation for the pathological changes observed in conditions such as pre-eclampsia (PE).
Collapse
Affiliation(s)
| | - Carmela Rianna
- Institute of Applied Physics, University of Tübingen, Tübingen, Germany
| | - Emily Hellwich
- Institute of Applied Physics, University of Tübingen, Tübingen, Germany
| | - Iliana Nikolou
- Department of Women's Health, University of Tübingen, Tübingen, Germany
| | | | - Sara Y Brucker
- Department of Women's Health, University of Tübingen, Tübingen, Germany
| | - Yogesh Singh
- Department of Women's Health, University of Tübingen, Tübingen, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Tilman E Schäffer
- Institute of Applied Physics, University of Tübingen, Tübingen, Germany
| | - Madhuri S Salker
- Department of Women's Health, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
106
|
Papastefanou I, Menenez M, Szczepkowska A, Gungil B, Syngelaki A, Nicolaides KH. Comparison of competing-risks model with angiogenic factors in midgestation screening for preterm growth-related neonatal morbidity. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2024; 63:613-618. [PMID: 38057964 DOI: 10.1002/uog.27559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023]
Abstract
OBJECTIVES First, to evaluate the predictive performance for preterm growth-related neonatal morbidity of a high soluble fms-like tyrosine kinase-1 (sFlt-1)/placental growth factor (PlGF) ratio or low PlGF at midgestation and, second, to compare the performance of a high sFlt-1/PlGF ratio or low PlGF with that of the competing-risks model for small-for-gestational age (SGA), utilizing a combination of maternal risk factors, sonographic estimated fetal weight and uterine artery pulsatility index. METHODS This was a prospective observational study in women attending for a routine hospital visit at 19-24 weeks' gestation in two maternity hospitals in England. The visit included recording of maternal demographic characteristics and medical history, carrying out an ultrasound scan and measuring serum PlGF and sFlt-1. The primary outcome was delivery < 32 and < 37 weeks' gestation of a SGA neonate with birth weight < 10th or < 3rd percentile, combined with neonatal unit (NNU) admission for ≥ 48 h or a composite of major neonatal morbidity. The detection rates in screening by PlGF < 10th percentile, sFlt-1/PlGF ratio > 90th percentile and the competing-risks model for SGA were estimated and then compared using McNemar's test. RESULTS In the study population of 40 241 women, prediction of preterm growth-related neonatal morbidity provided by the competing-risks model for SGA was superior to that of screening by low PlGF concentration or high sFlt-1/PlGF ratio. For example, at a screen-positive rate of 10.0%, as defined by the sFlt-1/PlGF ratio > 90th percentile, the competing-risks model predicted 70.1% (95% CI, 61.0-79.2%) of SGA < 10th percentile and 76.9% (95% CI, 67.6-86.3%) of SGA < 3rd percentile with NNU admission for ≥ 48 h delivered < 32 weeks' gestation. The respective values for SGA with major neonatal morbidity were 73.8% (95% CI, 64.4-83.2%) and 77.9% (95% CI, 68.0-87.8%). These were significantly higher than the respective values of 35.1% (95% CI, 25.6-44.6%), 35.9% (95% CI, 25.3-46.5%), 38.1% (95% CI, 27.7-48.5%) and 39.7% (95% CI, 28.1-51.3%) achieved by the application of the sFlt-1/PlGF ratio > 90th percentile (all P < 0.0001). CONCLUSION At midgestation, the prediction of growth-related neonatal morbidity by the competing-risks model for SGA is superior to that of a high sFlt-1/PlGF ratio or low PlGF. © 2023 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- I Papastefanou
- Department of Women and Children's Health, School of Life Course and Population Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - M Menenez
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - A Szczepkowska
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - B Gungil
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - A Syngelaki
- Department of Women and Children's Health, School of Life Course and Population Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - K H Nicolaides
- Department of Women and Children's Health, School of Life Course and Population Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| |
Collapse
|
107
|
Callbo PN, Junus K, Gabrysch K, Bergman L, Poromaa IS, Lager S, Wikström AK. Novel Associations Between Mid-Pregnancy Cardiovascular Biomarkers and Preeclampsia: An Explorative Nested Case-Control Study. Reprod Sci 2024; 31:1391-1400. [PMID: 38253981 PMCID: PMC11090924 DOI: 10.1007/s43032-023-01445-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: 05/13/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024]
Abstract
Prediction of women at high risk of preeclampsia is important for prevention and increased surveillance of the disease. Current prediction models need improvement, particularly with regard to late-onset preeclampsia. Preeclampsia shares pathophysiological entities with cardiovascular disease; thus, cardiovascular biomarkers may contribute to improving prediction models. In this nested case-control study, we explored the predictive importance of mid-pregnancy cardiovascular biomarkers for subsequent preeclampsia. We included healthy women with singleton pregnancies who had donated blood in mid-pregnancy (~ 18 weeks' gestation). Cases were women with subsequent preeclampsia (n = 296, 10% of whom had early-onset preeclampsia [< 34 weeks]). Controls were women who had healthy pregnancies (n = 333). We collected data on maternal, pregnancy, and infant characteristics from medical records. We used the Olink cardiovascular II panel immunoassay to measure 92 biomarkers in the mid-pregnancy plasma samples. The Boruta algorithm was used to determine the predictive importance of the investigated biomarkers and first-trimester pregnancy characteristics for the development of preeclampsia. The following biomarkers had confirmed associations with early-onset preeclampsia (in descending order of importance): placental growth factor (PlGF), matrix metalloproteinase (MMP-12), lectin-like oxidized LDL receptor 1, carcinoembryonic antigen-related cell adhesion molecule 8, serine protease 27, pro-interleukin-16, and poly (ADP-ribose) polymerase 1. The biomarkers that were associated with late-onset preeclampsia were BNP, MMP-12, alpha-L-iduronidase (IDUA), PlGF, low-affinity immunoglobulin gamma Fc region receptor II-b, and T cell surface glycoprotein. Our results suggest that MMP-12 is a promising novel preeclampsia biomarker. Moreover, BNP and IDUA may be of value in enhancing prediction of late-onset preeclampsia.
Collapse
Affiliation(s)
- Paliz Nordlöf Callbo
- Department of Women's and Children's Health, Uppsala University, Akademiska sjukhuset, SE 751 85, Uppsala, Sweden.
| | - Katja Junus
- Department of Women's and Children's Health, Uppsala University, Akademiska sjukhuset, SE 751 85, Uppsala, Sweden
| | | | - Lina Bergman
- Department of Women's and Children's Health, Uppsala University, Akademiska sjukhuset, SE 751 85, Uppsala, Sweden
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Obstetrics and Gynecology, Stellenbosch University, Cape Town, South Africa
| | - Inger Sundström Poromaa
- Department of Women's and Children's Health, Uppsala University, Akademiska sjukhuset, SE 751 85, Uppsala, Sweden
| | - Susanne Lager
- Department of Women's and Children's Health, Uppsala University, Akademiska sjukhuset, SE 751 85, Uppsala, Sweden
| | - Anna-Karin Wikström
- Department of Women's and Children's Health, Uppsala University, Akademiska sjukhuset, SE 751 85, Uppsala, Sweden
| |
Collapse
|
108
|
Januzzi JL, Liu Y, Sattar N, Yavin Y, Pollock CA, Butler J, Jardine M, Heerspink HJL, Masson S, Breyer M, Hansen MK. Vascular endothelial growth factors and risk of cardio-renal events: Results from the CREDENCE trial. Am Heart J 2024; 271:38-47. [PMID: 38401646 DOI: 10.1016/j.ahj.2024.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/30/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Circulating concentrations of vascular endothelial growth factor (VEGF) family members may be abnormally elevated in type 2 diabetes (T2D). The roles of placental growth factor (PlGF), soluble fms-like tyrosine kinase-1 (sFLT-1), and VEGF-A in cardio-renal complications of T2D are not established. METHOD The 2602 individuals with diabetic kidney disease (DKD) from the Canagliflozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation trial were randomized to receive canagliflozin or placebo and followed for incident cardio-renal outcomes. PlGF, sFLT-1, and VEGF-A were measured at baseline, year 1, and year 3. Primary outcome was a composite of end-stage kidney disease, doubling of the serum creatinine, or renal/cardiovascular death. Cox proportional hazard regression was used to investigate the association between biomarkers with adverse clinical events. RESULTS At baseline, individuals with higher PlGF levels had more prevalent cardiovascular disease compared to those with lower values. Treatment with canagliflozin did not meaningfully change PlGF, sFLT-1, and VEGF-A concentrations at years 1 and 3. In a multivariable model, 1 unit increases in baseline log PlGF (hazard ratio [HR]: 1.76, 95% confidence interval [CI]: 1.23, 2.54, P-value = .002), sFLT-1 (HR: 3.34, [95% CI: 1.71, 6.52], P-value < .001), and PlGF/sFLT-1 ratio (HR: 4.83, [95% CI: 0.86, 27.01], P-value = .07) were associated with primary composite outcome, while 1 unit increase in log VEGF-A did not increase the risk of primary outcome (HR: 0.96 [95% CI: 0.81, 1.07]). Change by 1 year of each biomarker was also assessed: HR (95% CI) of primary composite outcome was 2.45 (1.70, 3.54) for 1 unit increase in 1-year concentration of log PlGF, 4.19 (2.18, 8.03) for 1 unit increase in 1-year concentration of log sFLT-1, and 21.08 (3.79, 117.4) for 1 unit increase in 1-year concentration of log PlGF/sFLT-1. Increase in 1-year concentrations of log VEGF-A was not associated with primary composite outcome (HR: 1.08, [95% CI: 0.93, 1.24], P-value = .30). CONCLUSIONS People with T2D and DKD with elevated levels of PlGF, sFLT-1, and PlGF/sFLT-1 ratio were at a higher risk for cardiorenal events. Canagliflozin did not meaningfully decrease concentrations of PlGF, sFLT-1, and VEGF-A. CLINICAL TRIAL CREDENCE, https://clinicaltrials.gov/ct2/show/NCT02065791.
Collapse
Affiliation(s)
- James L Januzzi
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA; Heart Failure and Biomarker Trials, Baim Institute for Clinical Research, Boston, MA.
| | - Yuxi Liu
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Yshai Yavin
- Janssen Research & Development, LLC, Spring House, PA
| | - Carol A Pollock
- Kolling Institute, Royal North Shore Hospital University of Sydney, Sydney, NSW, Australia
| | - Javed Butler
- University of Mississippi Medical Center, Jackson, MS; Baylor Scott & White Institute, Dallas, TX
| | - Meg Jardine
- The George Institute for Global Health, UNSW Sydney, Sydney, NSW
| | - Hiddo J L Heerspink
- Department Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, The Netherlands
| | - Serge Masson
- Roche Diagnostics International, Rotkreuz, Switzerland
| | | | | |
Collapse
|
109
|
Palmrich P, Kalafat E, Pateisky P, Schirwani-Hartl N, Haberl C, Herrmann C, Khalil A, Binder J. Prognostic value of angiogenic markers in pregnancy with fetal growth restriction. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2024; 63:619-626. [PMID: 37774098 DOI: 10.1002/uog.27509] [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: 05/19/2023] [Revised: 09/01/2023] [Accepted: 09/20/2023] [Indexed: 10/01/2023]
Abstract
OBJECTIVE Pregnancies with fetal growth restriction (FGR) are at increased risk for pre-eclampsia. Angiogenic markers including soluble fms-like tyrosine kinase-1 (sFlt-1) and placental growth factor (PlGF) are altered in pregnancies complicated by FGR, but their utility for predicting pre-eclampsia in growth-restricted pregnancies is uncertain. This study aimed to evaluate the prognostic value of angiogenic markers for predicting the development of pre-eclampsia in pregnancies with FGR and suspected pre-eclampsia. METHODS This was a retrospective study of singleton pregnancies with FGR, defined according to Delphi consensus criteria, which underwent sampling of sFlt-1 and PlGF for suspicion of pre-eclampsia at the Medical University of Vienna, Vienna, Austria, between 2013 and 2020. Women with an established diagnosis of pre-eclampsia at sampling were excluded. Cox regression analysis and logistic regression analysis were performed to evaluate the association of angiogenic markers with the development of pre-eclampsia at various timepoints. RESULTS In this cohort of 93 women, pre-eclampsia was diagnosed in 14 (15.1%) women within 1 week after sampling, 21 (22.6%) within 2 weeks after sampling and 38 (40.9%) at any time after assessment. The sFlt-1/PlGF ratio consistently showed a stronger association with the development of pre-eclampsia compared to sFlt-1 or PlGF alone (pre-eclampsia within 1 week: area under the receiver-operating-characteristics curve, 0.87 vs 0.82 vs 0.72). Models including the sFlt-1/PlGF ratio were associated more strongly with pre-eclampsia hazard compared to models including sFlt-1 or PlGF alone (concordance index, 0.790 vs 0.759 vs 0.755). The risk classification capability of the sFlt-1/PlGF ratio decreased after the 2-week timepoint. The established cut-off value for the sFlt-1/PlGF ratio of < 38 was effective for ruling out pre-eclampsia within 2 weeks, with a negative predictive value of 0.933 and sensitivity of 0.952. CONCLUSIONS Use of the sFlt-1/PlGF ratio is preferrable to the use of PlGF alone for the prediction of pre-eclampsia in pregnancies with FGR. Established cut-offs for ruling out the development of pre-eclampsia in the short term seem to be effective in these patients. © 2023 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- P Palmrich
- Department of Obstetrics and Feto-Maternal Medicine, Medical University of Vienna, Vienna, Austria
| | - E Kalafat
- Department of Obstetrics and Gynecology, School of Medicine, Koc University, Istanbul, Turkey
| | - P Pateisky
- Department of Obstetrics and Feto-Maternal Medicine, Medical University of Vienna, Vienna, Austria
| | - N Schirwani-Hartl
- Department of Obstetrics and Feto-Maternal Medicine, Medical University of Vienna, Vienna, Austria
| | - C Haberl
- Department of Obstetrics and Feto-Maternal Medicine, Medical University of Vienna, Vienna, Austria
| | - C Herrmann
- Department of Obstetrics and Feto-Maternal Medicine, Medical University of Vienna, Vienna, Austria
| | - A Khalil
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust, University of London, London, UK
- Vascular Biology Research Centre, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - J Binder
- Department of Obstetrics and Feto-Maternal Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
110
|
Hong J, Crawford K, Cavanagh E, da Silva Costa F, Kumar S. Placental growth factor and fetoplacental Doppler indices in combination predict preterm birth reliably in pregnancies complicated by fetal growth restriction. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2024; 63:635-643. [PMID: 37820083 DOI: 10.1002/uog.27513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/25/2023] [Accepted: 10/02/2023] [Indexed: 10/13/2023]
Abstract
OBJECTIVE To assess the association between placental biomarkers (placental growth factor (PlGF) and soluble fms-like tyrosine kinase-1 (sFlt-1)/PlGF ratio) and fetoplacental Doppler indices (umbilical artery (UA) pulsatility index (PI) and uterine artery (UtA) PI) in various combinations for predicting preterm birth (PTB) in pregnancies complicated by fetal growth restriction (FGR). METHODS This was a prospective observational cohort study, performed at Mater Mother's Hospital in Brisbane, Queensland, Australia, from May 2022 to June 2023, of pregnancies complicated by FGR and appropriate-for-gestational-age (AGA) pregnancies. Maternal serum PlGF levels, sFlt-1/PlGF ratio, UA-PI and UtA-PI were measured at 2-4-weekly intervals from recruitment until delivery. Harrell's concordance statistic (Harrell's C) was used to evaluate multivariable Cox proportional hazards regression models featuring various combinations of placental biomarkers and fetoplacental Doppler indices to ascertain the best combination to predict PTB (< 37 weeks). Multivariable Cox regression models were used with biomarkers as time-varying covariates. RESULTS The study cohort included 320 singleton pregnancies, comprising 179 (55.9%) affected by FGR, defined according to a Delphi consensus, and 141 (44.1%) with an AGA fetus. In the FGR cohort, both low PlGF levels and elevated sFlt-1/PlGF ratio were associated with significantly shorter time to PTB. Low PlGF was a better predictor of PTB than was either sFlt-1/PlGF ratio or a combination of PlGF and sFlt-1/PlGF ratio (Harrell's C, 0.81, 0.78 and 0.79, respectively). Although both Doppler indices were significantly associated with time to PTB, in combination they were better predictors of PTB than was either UA-PI > 95th centile or UtA-PI > 95th centile alone (Harrell's C, 0.82, 0.75 and 0.76, respectively). Predictive utility for PTB was best when PlGF < 100 ng/L, UA-PI > 95th centile and UtA-PI > 95th centile were combined (Harrell's C, 0.88) (hazard ratio, 32.99; 95% CI, 10.74-101.32). CONCLUSIONS Low maternal serum PlGF level (< 100 ng/L) and abnormal fetoplacental Doppler indices (UA-PI > 95th centile and UtA-PI > 95th centile) in combination have the greatest predictive utility for PTB in pregnancies complicated by FGR. Their assessment may help guide clinical management of these complex pregnancies. © 2023 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- J Hong
- Mater Research Institute, University of Queensland, South Brisbane, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - K Crawford
- Mater Research Institute, University of Queensland, South Brisbane, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - E Cavanagh
- Mater Research Institute, University of Queensland, South Brisbane, Queensland, Australia
| | - F da Silva Costa
- School of Medicine and Dentistry, Griffith University and Maternal Fetal Medicine Unit, Gold Coast University Hospital, Gold Coast, Queensland, Australia
| | - S Kumar
- Mater Research Institute, University of Queensland, South Brisbane, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
- NHMRC Centre for Research Excellence in Stillbirth, Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
111
|
Izumi S, Iwama N, Metoki H. Prediction of preterm preeclampsia risk in Asians using a simple two-item assessment in early pregnancy. Hypertens Res 2024; 47:1231-1234. [PMID: 38321104 DOI: 10.1038/s41440-024-01590-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 12/30/2023] [Indexed: 02/08/2024]
Affiliation(s)
- Seiya Izumi
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, 1-1, Seiryo-machi, Sendai, 980-8574, Miyagi, Japan
| | - Noriyuki Iwama
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, 1-1, Seiryo-machi, Sendai, 980-8574, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Sendai, 980-8573, Miyagi, Japan
| | - Hirohito Metoki
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Sendai, 980-8573, Miyagi, Japan.
- Division of Public Health, Hygiene and Epidemiology, Tohoku Medical Pharmaceutical University, 1-15-1 Fukumuro, Sendai, 983-8536, Miyagi, Japan.
| |
Collapse
|
112
|
Melo P, Devall A, Shennan AH, Vatish M, Becker CM, Granne I, Papageorghiou AT, Mol BW, Coomarasamy A. Vaginal micronised progesterone for the prevention of hypertensive disorders of pregnancy: A systematic review and meta-analysis. BJOG 2024; 131:727-739. [PMID: 37941309 DOI: 10.1111/1471-0528.17705] [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: 06/30/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Treatment with vaginal progesterone reduces the risk of miscarriage and preterm birth in selected high-risk women. The hypothesis that vaginal progesterone can reduce the risk of hypertensive disorders of pregnancy (HDP) is unexplored. OBJECTIVES To summarise the evidence on the effectiveness of vaginal progesterone to reduce the risk of HDP. SEARCH STRATEGY We searched Embase (OVID), MEDLINE (OVID), PubMed, CENTRAL and clinicaltrials.gov from inception until 20 June 2023. SELECTION CRITERIA We included placebo-controlled randomised trials (RCTs) of vaginal progesterone for the prevention or treatment of any pregnancy complications. DATA COLLECTION AND ANALYSIS We extracted absolute event numbers for HDP and pre-eclampsia in women receiving vaginal progesterone or placebo, and meta-analysed the data with a random effects model. We appraised the certainty of the evidence using GRADE methodology. MAIN RESULTS The quantitative synthesis included 11 RCTs, of which three initiated vaginal progesterone in the first trimester, and eight in the second or third trimesters. Vaginal progesterone started in the first trimester of pregnancy lowered the risk of any HDP (risk ratio [RR] 0.71, 95% confidence interval [CI] 0.53-0.93, 2 RCTs, n = 4431 women, I2 = 0%; moderate-certainty evidence) and pre-eclampsia (RR 0.61, 95% CI 0.41-0.92, 3 RCTs, n = 5267 women, I2 = 0%; moderate-certainty evidence) when compared with placebo. Vaginal progesterone started in the second or third trimesters was not associated with a reduction in HDP (RR 1.19, 95% CI 0.67-2.12, 3 RCTs, n = 1602 women, I2 = 9%; low-certainty evidence) or pre-eclampsia (RR 0.97, 95% CI 0.71-1.31, 5 RCTs, n = 4274 women, I2 = 0%; low-certainty evidence). CONCLUSIONS Our systematic review found first-trimester initiated vaginal micronised progesterone may reduce the risk of HDP and pre-eclampsia.
Collapse
Affiliation(s)
- Pedro Melo
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
- Tommy's National Centre for Miscarriage Research, College of Medical and Dental Sciences, Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, UK
| | - Adam Devall
- Tommy's National Centre for Miscarriage Research, College of Medical and Dental Sciences, Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, UK
| | - Andrew H Shennan
- Department of Women and Children's Health, King's College London, London, UK
| | - Manu Vatish
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Christian M Becker
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Ingrid Granne
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Aris T Papageorghiou
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Ben W Mol
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
- Aberdeen Centre for Women's Health Research, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Arri Coomarasamy
- Tommy's National Centre for Miscarriage Research, College of Medical and Dental Sciences, Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, UK
| |
Collapse
|
113
|
Chirilă CN, Mărginean C, Ghiga DV, Voidăzan S, Chirilă PM, Gliga ML. A Second Trimester Prediction Algorithm for Early-Onset Hypertensive Disorders of Pregnancy Occurrence and Severity Based on Soluble fms-like Tyrosine Kinase 1 (sFlt-1)/Placental Growth Factor (PlGF) Ratio and Uterine Doppler Ultrasound in Women at Risk. CHILDREN (BASEL, SWITZERLAND) 2024; 11:468. [PMID: 38671685 PMCID: PMC11049313 DOI: 10.3390/children11040468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024]
Abstract
Hypertensive disorders of pregnancy (HDPs) represent a significant source of severe maternal and fetal morbidity. Screening strategies relying on traditional medical history and clinical risk factors have traditionally shown relatively modest performance, mainly in the prediction of preeclampsia, displaying a sensitivity of 37% for the early-onset form and 29% for the late-onset form. The development of more accurate predictive and diagnostic models of preeclampsia in the early stages of pregnancy represents a matter of high priority. The aim of the present paper is to create an effective second trimester prediction algorithm of early-onset HDP occurrence and severity, by combining the following two biochemical markers: a soluble fms-like tyrosine kinase 1 (sFlt-1)/placental growth factor (PlGF) ratio and uterine artery Doppler ultrasound parameters, namely the pulsatility index (PI) and the resistivity index (RI), in a population of high-risk pregnant women, initially assessed through traditional risk factors. A prospective single-center observational longitudinal study was conducted, in which 100 women with singleton pregnancy and traditional clinical and medical history risk factors for preeclampsia were enrolled at 24 weeks of gestation. Shortly after study enrollment, all women had their sFlt-1 and PlGF levels and mean uterine artery PI and RI determined. All pregnancies were followed up until delivery. Receiver operating characteristic (ROC) analysis established algorithms based on cutoffs for the prediction of the later development of preeclampsia: PI 1.25 (96.15% sensitivity, 86.49% specificity), RI 0.62 (84.6% sensitivity, 89.2% specificity) and sFlt-1/PlGF ratio 59.55 (100% sensitivity, 89.2% specificity). The sFlt-1/PlGF ratio was the best predictor for preeclampsia, as it displayed the highest area under the curve (AUC) of 0.973. The prediction algorithm for the severe form of preeclampsia, complicated by fetal growth restriction leading to preterm birth, antepartum fetal demise or acute fetal distress with a cerebro-placental ratio of
Collapse
Affiliation(s)
- Cristian Nicolae Chirilă
- Department of Internal Medicine-Nephrology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (C.N.C.); (M.L.G.)
- Department of Nephrology, Mures Clinical County Hospital, 540103 Târgu Mureș, Romania
- Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Claudiu Mărginean
- Department of Obstetrics and Gynecology 2, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania
- Department of Obstetrics and Gynecology, Mures Clinical County Hospital, 540057 Târgu Mureș, Romania
| | - Dana Valentina Ghiga
- Department of Scientific Medical Research Methodology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Septimiu Voidăzan
- Department of Epidemiology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Paula Maria Chirilă
- Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania;
- Department of Endocrinology, Mures Clinical County Hospital, 540142 Târgu Mureș, Romania
| | - Mirela Liana Gliga
- Department of Internal Medicine-Nephrology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (C.N.C.); (M.L.G.)
- Department of Nephrology, Mures Clinical County Hospital, 540103 Târgu Mureș, Romania
| |
Collapse
|
114
|
Choi J, Lee SM, Norwitz ER, Kim JH, Jung YM, Park CW, Jun JK, Lee D, Jin Y, Kim S, Cha B, Park JS, Kim JI. Placental expression quantitative trait loci in an East Asian population. HGG ADVANCES 2024; 5:100276. [PMID: 38310352 PMCID: PMC10883826 DOI: 10.1016/j.xhgg.2024.100276] [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: 08/11/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024] Open
Abstract
Expression quantitative trait loci (eQTL) analysis measures the contribution of genetic variation in gene expression on complex traits. Although this methodology has been used to examine gene regulation in numerous human tissues, eQTL research in solid tissues is relatively lacking. We conducted eQTL analysis on placentas collected from an East Asian population in an effort to identify gene regulatory mechanisms in this tissue. Placentas (n = 102) were collected at the time of cesarean delivery. mRNA was extracted, sequenced with NGS, and compared with matched maternal and fetal DNA arrays performed using maternal and neonatal cord blood. Linear regression modeling was performed using tensorQTL. Fine-mapping along with epigenomic annotation was used to select putative functional variants. We identified 2,703 coding genes that contained at least one eQTL with statistical significance (false discovery rate <0.05). After fine-mapping, we found 108 previously unreported eQTL variants with posterior inclusion probability >0.1. Of these, 19% were located in genomic regions with evidence from public placental epigenome suggesting that they may be functionally relevant. For example, variant rs28379289 located in the placenta-specific regulatory region changes the binding affinity of transcription factor leading to higher expression of LGALS3, which is known to affect placental function. This study expands the knowledge base of regulatory elements within the human placenta and identifies 108 previously unreported placenta eQTL signals, which are listed in our publicly available GMI eQTL database. Further studies are needed to identify and characterize genetic regulatory mechanisms that affect placental function in normal pregnancy and placenta-related diseases.
Collapse
Affiliation(s)
- Jaeyong Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea; Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, Korea
| | - Seung Mi Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | | | - Ji Hoi Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Young Mi Jung
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Chan-Wook Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Jong Kwan Jun
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Dakyung Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea; Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, Korea
| | - Yongjoon Jin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea; Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, Korea
| | - Sookyung Kim
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, Korea
| | - Bukyoung Cha
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, Korea
| | - Joong Shin Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea.
| | - Jong-Il Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea; Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, Korea.
| |
Collapse
|
115
|
Awoyemi T, Jiang S, Rahbar M, Logentherian P, Collett G, Zhang W, Cribbs A, Cerdeira S, Vatish M. MicroRNA analysis of medium/large placenta extracellular vesicles in normal and preeclampsia pregnancies. Front Cardiovasc Med 2024; 11:1371168. [PMID: 38628314 PMCID: PMC11018924 DOI: 10.3389/fcvm.2024.1371168] [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: 01/16/2024] [Accepted: 03/12/2024] [Indexed: 04/19/2024] Open
Abstract
Background Preeclampsia (PE) is a hypertensive disorder of pregnancy, affecting 2%-8% of pregnancies worldwide, and is the leading cause of adverse maternal and fetal outcomes. The disease is characterized by oxidative and cellular stress and widespread endothelial dysfunction. While the precise mechanisms are not entirely understood, the pathogenesis of PE is closely linked to placental dysfunction and, to some extent, syncytiotrophoblast extracellular vesicle release (STB-EVs). These vesicles can be divided into the less well-studied medium/large EVs (220-1,000 nm) released in response to stress and small EVs (<220 nm) released as a component of intercellular communication. The previously described production of m/lSTB-EVs in response to cellular stress combined with the overwhelming occurrence of cellular and oxidative stress in PE prompted us to evaluate the microRNAome of PE m/lSTB-EVs. We hypothesized that the microRNAome profile of m/lSTB-EVs is different in PE compared to normal pregnancy (NP), which might permit the identification of potential circulating biomarkers not previously described in PE. Methods/study design We performed small RNA sequencing on medium/large STB-EVs isolated from PE and NP placentae using dual-lobe ex vivo perfusion. The sequencing data was bioinformatically analyzed to identify differentially regulated microRNAs. Identified microRNAs were validated with quantitative PCR analysis. We completed our analysis by performing an in-silico prediction of STB-EV mechanistic pathways. Results We identified significant differences between PE and NP in the STB-EVs micro ribonucleic acid (microRNA) profiles. We verified the differential expression of hsa-miR-193b-5p, hsa-miR-324-5p, hsa-miR-652-3p, hsa-miR-3196, hsa-miR-9-5p, hsa-miR-421, and hsa-miR-210-3p in the medium/large STB-EVs. We also confirmed the differential abundance of hsa-miR-9-5p in maternal serum extracellular vesicles (S EVs). In addition, we integrated the results of these microRNAs into the previously published messenger RNA (mRNA) data to better understand the relationship between these biomolecules. Conclusions We identified a differentially regulated micro-RNA, hsa-miR-9-5p, that may have biomarker potential and uncovered mechanistic pathways that may be important in the pathophysiology of PE.
Collapse
Affiliation(s)
- Toluwalase Awoyemi
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Shuhan Jiang
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Maryam Rahbar
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Prasanna Logentherian
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Gavin Collett
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Wei Zhang
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Adam Cribbs
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Sofia Cerdeira
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Manu Vatish
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
116
|
Dionisio LM, Favero GM. Platelet indices and angiogenesis markers in hypertensive disorders of pregnancy. Int J Lab Hematol 2024; 46:259-265. [PMID: 37953406 DOI: 10.1111/ijlh.14202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/20/2023] [Indexed: 11/14/2023]
Abstract
INTRODUCTION Activated platelets exert a key role in the pathogenesis of preeclampsia (PE). There is evidence of distinctive patterns of platelet indices in PE in comparison to healthy pregnancies, therefore these indices can be potential tools for PE detection, risk stratification, and management. Considering the vascular aspects of its pathophysiology, PE is characterized by the increased levels of soluble FMS-like tyrosine kinase-1 (sFlt-1) an antiangiogenic factor, and reduced placental growth factor (PlGF), a proangiogenic factor. This study aimed to assess the platelet indices in hypertensive disorders of pregnancy (HDP) and its correlation with angiogenesis-related biomarkers. METHODS The groups for the study were: control (n = 114); gestational hypertension; (n = 112), and PE (n = 42). The platelet indices included were platelet counts (PLT-I and PLT-F), mean platelet volume (MPV), platelet distribution width (PDW), plateletcrit (PCT), platelet large cell ratio (P-LCR), and immature platelet fraction (IPF# and IPF%). Serum levels of sFlt-1 and PlGF were assessed. RESULTS PLT-I, PLT-F, and PCT% were lower in PE, while MPV, PDW, P-LCR, IPF%, and IPF# were increased. The parameter MPV presented the best performance for the discrimination of PE. There was a moderate positive correlation between sFlt-1 levels and MPV, PDW, and P-LCR. CONCLUSION Platelet indices can be potentially applied as additional tools for the diagnosis and management of HDP. Activated platelets may act as an extra source of sFlt-1 in PE.
Collapse
Affiliation(s)
- Laura Mattana Dionisio
- Department of Clinical Analysis and Toxicology, State University of Ponta Grossa, Ponta Grossa, Parana, Brazil
- Department of General Biology, State University of Ponta Grossa, Ponta Grossa, Parana, Brazil
| | - Giovani Marino Favero
- Department of Clinical Analysis and Toxicology, State University of Ponta Grossa, Ponta Grossa, Parana, Brazil
- Department of General Biology, State University of Ponta Grossa, Ponta Grossa, Parana, Brazil
| |
Collapse
|
117
|
Burwick RM, Rodriguez MH. Angiogenic Biomarkers in Preeclampsia. Obstet Gynecol 2024; 143:515-523. [PMID: 38350106 DOI: 10.1097/aog.0000000000005532] [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: 11/07/2023] [Accepted: 01/04/2024] [Indexed: 02/15/2024]
Abstract
Preeclampsia contributes disproportionately to maternal and neonatal morbidity and mortality throughout the world. A critical driver of preeclampsia is angiogenic imbalance, which is often present weeks to months before overt disease. Two placenta-derived angiogenic biomarkers, soluble fms-like tyrosine kinase 1 (sFlt-1) and placental growth factor (PlGF), have proved useful as diagnostic and prognostic tests for preeclampsia. Recently, the U.S. Food and Drug Administration approved the sFlt-1/PlGF assay to aid in the prediction of preeclampsia with severe features among women with hypertensive disorders of pregnancy at 24-34 weeks of gestation. In this narrative review, we summarize the body of work leading to this approval and describe how the sFlt-1/PlGF ratio may be implemented in clinical practice as an adjunctive measure to help optimize care and to reduce adverse outcomes in preeclampsia.
Collapse
Affiliation(s)
- Richard M Burwick
- Division of Maternal Fetal Medicine, San Gabriel Valley Perinatal Medical Group, Pomona Valley Hospital Medical Center, Pomona, California
| | | |
Collapse
|
118
|
CHAIWORAPONGSA T, ROMERO R, GOMEZ-LOPEZ N, SUKSAI M, GALLO DM, JUNG E, BERRY SM, AWONUGA A, TARCA AL, BRYANT DR. Preeclampsia at term: evidence of disease heterogeneity based on the profile of circulating cytokines and angiogenic factors. Am J Obstet Gynecol 2024; 230:450.e1-450.e18. [PMID: 37806612 PMCID: PMC10990810 DOI: 10.1016/j.ajog.2023.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Intravascular inflammation and an antiangiogenic state have been implicated in the pathophysiology of preeclampsia. On the basis of the profiles of their angiogenic/antiangiogenic factors, women with preeclampsia at term may be classified into 2 subgroups with different characteristics and prevalence of adverse outcomes. This study was undertaken to examine whether these 2 subgroups of preeclampsia at term also show differences in their profiles of intravascular inflammation. OBJECTIVE This study aimed to determine the plasma profiles of cytokines and chemokines in women with preeclampsia at term who had a normal or an abnormal angiogenic profile. STUDY DESIGN A nested case-control study was conducted to include women classified into 3 groups: women with an uncomplicated pregnancy (n=213) and women with preeclampsia at term with a normal (n=55) or an abnormal (n=41) angiogenic profile. An abnormal angiogenic profile was defined as a plasma ratio of placental growth factor and soluble fms-like tyrosine kinase-1 multiple of the median <10th percentile for gestational age. Concentrations of cytokines were measured by multiplex immunoassays. RESULTS Women with preeclampsia at term and an abnormal angiogenic profile showed evidence of the greatest intravascular inflammation among the study groups. These women had higher plasma concentrations of 5 cytokines (interleukin-6, interleukin-8, interleukin-12/interleukin-23p40, interleukin-15, and interleukin-16) and 7 chemokines (eotaxin, eotaxin-3, interferon-γ inducible protein-10, monocyte chemotactic protein-4, macrophage inflammatory protein-1β, macrophage-derived chemokine, and thymus and activation-regulated chemokine compared to women with an uncomplicated pregnancy. By contrast, women with preeclampsia at term and a normal angiogenic profile, compared to women with an uncomplicated pregnancy, had only a higher plasma concentration of monocyte chemotactic protein-4. A correlation between severity of the antiangiogenic state, blood pressure, and plasma concentrations of a subset of cytokines was observed. CONCLUSION Term preeclampsia can be classified into 2 clusters. One is characterized by an antiangiogenic state coupled with an excessive inflammatory process, whereas the other has neither of these features. These findings further support the heterogeneity of preeclampsia at term and may explain the distinct clinical outcomes.
Collapse
Affiliation(s)
- Tinnakorn CHAIWORAPONGSA
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto ROMERO
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
| | - Nardhy GOMEZ-LOPEZ
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Manaphat SUKSAI
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Dahiana M. GALLO
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Valle, Cali, Colombia
| | - Eunjung JUNG
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Stanley M. BERRY
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Awoniyi AWONUGA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Adi L. TARCA
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, USA
| | - David R. BRYANT
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
119
|
Chen Y, Di X, Xiang G, Liu Y, Pan X, Deng W, Zhu X, Lei M, Zhang G, Liu H. sFlt-1/PIGF ratio positive associated with non-dipper type change in ambulatory blood pressure monitoring(ABPM) for preeclampsia development. Hypertens Res 2024; 47:849-858. [PMID: 38017185 PMCID: PMC10994835 DOI: 10.1038/s41440-023-01509-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 10/13/2023] [Accepted: 10/20/2023] [Indexed: 11/30/2023]
Abstract
In order to explore relationship of ambulatory blood pressure monitoring (ABPM) and soluble fms-like tyrosine kinase-1/placental growth factor (sFlt-1/PlGF) in suspected preeclampsia(PE), suspected PE participants in 28 + 0 to 33 + 6 weeks underwent ABPM and sFlt-1/PlGF from July 2020 to July 2022 were included(N = 476) in study. ABPM parameters were compared between sFlt-1/PlGF ≥38 and <38 groups. Correlation analysis was performed between ABPM and sFlt-1/PlGF, and logistic regression was used to explore prediction value for PE in 2 weeks. One hundred eighteen cases developed PE in 2 weeks with 114 from sFlt-1/PlGF ≥38 group. Daytime and nighttime BP were all increased,with increased non-dipper (58.4% vs. 30.3%), riser (22.1% vs. 13.1%) and and decreased Dipper (15.4% vs. 45.9%) type of ABPM in sFlt-1/PlGF ≥38 groups (P < 0.05).The riser group had the highest sFlt-1 and lowest PlGF. sFlt-1/PlGF and sFlt-1 were all positively correlated with systolic (SBP) & diastolic blood pressure(DBP)(P < 0.01), in which correlation coefficients of daytime and nighttime BP with sFlt-1 were β = 150.05 & 157.67 for SBP, β = 234 and 199.01 for DBP, respectively. However, PlGF was only negatively associated with nighttime SBP and DBP(P < 0.05), with no correlation with daytime BP (P > 0.05).Combining sFlt-1/PlGF and ABPM model, showed sFlt-1/PlGF (aOR = 2.01 (1.69-2.36)), Nighttime DBP (aOR = 1.14 (1.02-1.28)) contributed to preeclampsia prediction, and had improved predictive value compared to ABPM or sFlt-1/PlGF models alone(P < 0.05). sFlt-1/PlGF ratio was positively correlated with BP parameters, whereas PIGF was only negatively correlated with nocturnal BP and increased non-dipper type change in ABPM, which had a synergistic effect with sFlt-1/PlGF on PE prediction.
Collapse
Affiliation(s)
- Yunshan Chen
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiaodan Di
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Guochun Xiang
- School of Health Management, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yunfeng Liu
- Electrocardiographic Monitoring Unit, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 9 Jinsui Road, Guangzhou, 510623, China
| | - Xiuyu Pan
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wenfeng Deng
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiongjie Zhu
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ming Lei
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Guozheng Zhang
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Huishu Liu
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
120
|
Motomura K, Morita H, Naruse K, Saito H, Matsumoto K. Implication of viruses in the etiology of preeclampsia. Am J Reprod Immunol 2024; 91:e13844. [PMID: 38627916 DOI: 10.1111/aji.13844] [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: 11/29/2023] [Revised: 03/12/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024] Open
Abstract
Preeclampsia is one of the most common disorders that poses threat to both mothers and neonates and a major contributor to perinatal morbidity and mortality worldwide. Viral infection during pregnancy is not typically considered to cause preeclampsia; however, syndromic nature of preeclampsia etiology and the immunomodulatory effects of viral infections suggest that microbes could trigger a subset of preeclampsia. Notably, SARS-CoV-2 infection is associated with an increased risk of preeclampsia. Herein, we review the potential role of viral infections in this great obstetrical syndrome. According to in vitro and in vivo experimental studies, viral infections can cause preeclampsia by introducing poor placentation, syncytiotrophoblast stress, and/or maternal systemic inflammation, which are all known to play a critical role in the development of preeclampsia. Moreover, clinical and experimental investigations have suggested a link between several viruses and the onset of preeclampsia via multiple pathways. However, the results of experimental and clinical research are not always consistent. Therefore, future studies should investigate the causal link between viral infections and preeclampsia to elucidate the mechanism behind this relationship and the etiology of preeclampsia itself.
Collapse
Affiliation(s)
- Kenichiro Motomura
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
- Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Hideaki Morita
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan
| | - Katsuhiko Naruse
- Department of Obstetrics and Gynecology, Dokkyo Medical University, Tochigi, Japan
| | - Hirohisa Saito
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
121
|
Püschl IC, Thaneswaran Vyramuthu M, Bonde L, Lebech M, Iraqi Møller H, Vauvert F Hviid T, Lund Sørensen B, Macklon NS. Is salivary uric acid, a putative biomarker of pre-eclampsia, of maternal, placental, or fetal origin? Eur J Obstet Gynecol Reprod Biol 2024; 295:34-41. [PMID: 38330864 DOI: 10.1016/j.ejogrb.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 01/29/2024] [Accepted: 02/02/2024] [Indexed: 02/10/2024]
Abstract
OBJECTIVES Increased salivary uric acid (sUA) represents a potential biomarker predictive of pre-eclampsia (PE), but its origin is unclear. The study explores whether sUA levels reflect maternal or feto-placental physiological stress and whether sUA levels in these cases correlate with amniotic fluid (fetal origin), maternal blood (maternal origin), or cord blood (fetal vs placental origin). STUDY DESIGN Pregnant women (n = 39) undergoing amniotomy or caesarean section after 34 gestational weeks were designated into three groups of either maternal, feto-placental, or no signs of physiological stress: women (n = 15) in the established first phase of active labour and without any signs of fetal growth restriction (FGR) or PE were assigned to the maternal stress group, women (n = 6) with an ultrasound-based diagnosis of FGR, with or without PE, were assigned to the feto-placental stress group, and women (n = 18) not yet in active labour and without any signs of FGR or PE, were assigned to the control group. Uric acid levels in corresponding samples of amniotic fluid, saliva, maternal blood, and cord blood were compared between groups and between body compartments within each group. RESULTS The feto-placental stress group showed increased UA levels in saliva (median, interquartile range [IQR]: 0.47 [0.38] mmol/L, P = 0.023) and maternal blood (0.42 [0.13] mmol/L, P = 0.032), but no differences in amniotic fluid or cord blood compared with the other groups. Within the control and maternal stress group, sUA levels were lower compared with maternal blood (0.20 [0.08] vs 0.25 [0.08] mmol/L, Pcontrol = 0.018; 0.20 [0.06] vs 0.26 [0.08] mmol/L, Pmaternal = 0.001) and highest in amniotic fluid (control group (0.49 [0.18] mmol/L): Pmaternal,blood = 0.001, Pumbilical,artery = <0.001, Pumbilical,vein = <0.001, Psaliva = <0.001) (maternal stress group (0.56 [0.23] mmol/L): Pmaternal,blood = 0.021, Pumbilical,artery = 0.006, Pumbilical,vein = 0.004, Psaliva = 0.003). Levels did not differ between compartments in the feto-placental stress group. CONCLUSIONS Salivary and maternal blood UA levels were increased in the feto-placental stress group with salivary levels increasing more than blood levels compared with the maternal stress and control groups, whilst UA in amniotic fluid were not different between the groups, suggesting a placental origin and potential use of sUA as a biomarker of placental dysfunction, including FGR and severe PE.
Collapse
Affiliation(s)
- Ida Catharina Püschl
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; Department of Obstetrics and Gynaecology and ReproHealth Research Consortium, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark.
| | - Meera Thaneswaran Vyramuthu
- Department of Obstetrics and Gynaecology and ReproHealth Research Consortium, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark
| | - Lisbeth Bonde
- Department of Obstetrics and Gynaecology, Juliane Marie Centre, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Morten Lebech
- Department of Obstetrics and Gynaecology and ReproHealth Research Consortium, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark
| | - Hiba Iraqi Møller
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; Department of Clinical Biochemistry and ReproHealth Research Consortium, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark
| | - Thomas Vauvert F Hviid
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; Department of Clinical Biochemistry and ReproHealth Research Consortium, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark
| | - Bjarke Lund Sørensen
- Department of Obstetrics and Gynaecology and ReproHealth Research Consortium, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark
| | - Nicholas S Macklon
- Department of Obstetrics and Gynaecology and ReproHealth Research Consortium, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark; London Women's Clinic, 113-115 Harley St, London W1G 6AP, United Kingdom
| |
Collapse
|
122
|
Wang L, Gao J, Tang P, Hu H, Chen X, Chen Z, Sun Y. Comparing urine point-of-care tests to screen preeclampsia: Congo-red dot paper test versus dipstick urinalysis. J Clin Hypertens (Greenwich) 2024; 26:349-354. [PMID: 38430477 PMCID: PMC11007796 DOI: 10.1111/jch.14783] [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: 12/02/2023] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 03/03/2024]
Abstract
To compare the urine Congo-red dot paper test (CRD) with dipstick urinalysis to screen preeclampsia (PE). A total of 409 paired spot urine samples were obtained prospectively from women with suspected pre-eclampsia attending for routine hospital visits. Congo-red dot paper test and dipstick urinalysis were examined and compared to screen pre-eclampsia. The agreement between the two urinary test is modest (kappa coefficient = 0.28, 95% CI 0.14-0.42). The specificity of CRD was higher than urinalysis (97.4% vs. 90.4%, p < .001). Urinalysis performed better in sensitivity (77.3% vs. 40.9%, p = .04) and the area under the receiver operating characteristic curves (AUC) (0.84 [95% CI 0.74-0.94] vs. 0.69 [95% CI 0.55-0.83], p = .04) than CRD, respectively. The sensitivity, specificity, AUC of the parallel test of them is 86.4% (64.0%-96.4%), 89.1% (85.5%-92.0%), and 0.88 (95% CI 0.79-0.96). And the serial test is 31.8% (14.7%-54.9%), 98.7% (96.8%-99.5%), 0.65 (95% CI 0.51-0.79), accordingly. The urinalysis is a better diagnosing test for preeclampsia. CRD could aid in the diagnosis of patients with preeclampsia. Combined the two tests in suspected patients may further improve the performance in the diagnosis of preeclampsia. Further study need to be made for its potential clinical practice.
Collapse
Affiliation(s)
- Liying Wang
- Department of Obstetrics and GynecologyChinese Academy of Medical SciencesPeking Union Medical CollegeNational Clinical Research Center for Obstetric and Gynecologic DiseasesPeking Union Medical College HospitalBeijingChina
| | - Jinsong Gao
- Department of Obstetrics and GynecologyChinese Academy of Medical SciencesPeking Union Medical CollegeNational Clinical Research Center for Obstetric and Gynecologic DiseasesPeking Union Medical College HospitalBeijingChina
| | - Pingping Tang
- Department of Obstetrics and GynecologyChinese Academy of Medical SciencesPeking Union Medical CollegeNational Clinical Research Center for Obstetric and Gynecologic DiseasesPeking Union Medical College HospitalBeijingChina
| | - Huiying Hu
- Department of Obstetrics and GynecologyChinese Academy of Medical SciencesPeking Union Medical CollegeNational Clinical Research Center for Obstetric and Gynecologic DiseasesPeking Union Medical College HospitalBeijingChina
| | - Xiaoxu Chen
- Department of Obstetrics and GynecologyChinese Academy of Medical SciencesPeking Union Medical CollegeNational Clinical Research Center for Obstetric and Gynecologic DiseasesPeking Union Medical College HospitalBeijingChina
| | - Ziyi Chen
- Department of Obstetrics and GynecologyChinese Academy of Medical SciencesPeking Union Medical CollegeNational Clinical Research Center for Obstetric and Gynecologic DiseasesPeking Union Medical College HospitalBeijingChina
| | - Yin Sun
- Department of Obstetrics and GynecologyChinese Academy of Medical SciencesPeking Union Medical CollegeNational Clinical Research Center for Obstetric and Gynecologic DiseasesPeking Union Medical College HospitalBeijingChina
| |
Collapse
|
123
|
Young RE, Nelson KM, Hofbauer SI, Vijayakumar T, Alameh MG, Weissman D, Papachristou C, Gleghorn JP, Riley RS. Systematic development of ionizable lipid nanoparticles for placental mRNA delivery using a design of experiments approach. Bioact Mater 2024; 34:125-137. [PMID: 38223537 PMCID: PMC10784148 DOI: 10.1016/j.bioactmat.2023.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 11/20/2023] [Accepted: 11/20/2023] [Indexed: 01/16/2024] Open
Abstract
Ionizable lipid nanoparticles (LNPs) have gained attention as mRNA delivery platforms for vaccination against COVID-19 and for protein replacement therapies. LNPs enhance mRNA stability, circulation time, cellular uptake, and preferential delivery to specific tissues compared to mRNA with no carrier platform. However, LNPs are only in the beginning stages of development for safe and effective mRNA delivery to the placenta to treat placental dysfunction. Here, we develop LNPs that enable high levels of mRNA delivery to trophoblasts in vitro and to the placenta in vivo with no toxicity. We conducted a Design of Experiments to explore how LNP composition, including the type and molar ratio of each lipid component, drives trophoblast and placental delivery. Our data revealed that utilizing C12-200 as the ionizable lipid and 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) as the phospholipid in the LNP design yields high transfection efficiency in vitro. Analysis of lipid molar composition as a design parameter in LNPs displayed a strong correlation between apparent pKa and poly (ethylene) glycol (PEG) content, as a reduction in PEG molar amount increases apparent pKa. Further, we present one LNP platform that exhibits the highest delivery of placental growth factor mRNA to the placenta in pregnant mice, resulting in synthesis and secretion of a potentially therapeutic protein. Lastly, our high-performing LNPs have no toxicity to both the pregnant mice and fetuses. Our results demonstrate the feasibility of LNPs as a platform for mRNA delivery to the placenta, and our top LNP formulations may provide a therapeutic platform to treat diseases that originate from placental dysfunction during pregnancy.
Collapse
Affiliation(s)
- Rachel E. Young
- Department of Biomedical Engineering, Henry M. Rowan College of Engineering, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States
- School of Translational Biomedical Engineering & Sciences, Virtua College of Medicine & Life Sciences of Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States
| | - Katherine M. Nelson
- Department of Chemical and Biomolecular Engineering, College of Engineering, University of Delaware, 150 Academy Street, Newark, DE 19716, United States
| | - Samuel I. Hofbauer
- Department of Biomedical Engineering, Henry M. Rowan College of Engineering, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States
- School of Translational Biomedical Engineering & Sciences, Virtua College of Medicine & Life Sciences of Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States
- Cooper Medical School of Rowan University, Rowan University, 401 Broadway, Camden, NJ 08103, United States
| | - Tara Vijayakumar
- Department of Biomedical Engineering, Henry M. Rowan College of Engineering, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States
- School of Translational Biomedical Engineering & Sciences, Virtua College of Medicine & Life Sciences of Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States
| | - Mohamad-Gabriel Alameh
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, United States
| | - Drew Weissman
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, United States
| | - Charalampos Papachristou
- Department of Mathematics, College of Science & Mathematics, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States
| | - Jason P. Gleghorn
- Department of Biomedical Engineering, College of Engineering, University of Delaware, 590 Avenue 1743, Newark, DE 19713, United States
| | - Rachel S. Riley
- Department of Biomedical Engineering, Henry M. Rowan College of Engineering, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States
- School of Translational Biomedical Engineering & Sciences, Virtua College of Medicine & Life Sciences of Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States
| |
Collapse
|
124
|
Tan L, Kluivers AC, Cruz-López EO, Broekhuizen M, Chen Z, Neuman RI, Schoenmakers S, Ruijgrok L, van de Velde D, de Winter BC, van den Bogaerdt AJ, Lu X, Danser AJ, Verdonk K. Statins Prevent the Deleterious Consequences of Placental Chemerin Upregulation in Preeclampsia. Hypertension 2024; 81:861-875. [PMID: 38361240 PMCID: PMC10956680 DOI: 10.1161/hypertensionaha.123.22457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/31/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND Chemerin, an inflammatory adipokine, is upregulated in preeclampsia, and its placental overexpression results in preeclampsia-like symptoms in mice. Statins may lower chemerin. METHODS Chemerin was determined in a prospective cohort study in women suspected of preeclampsia and evaluated as a predictor versus the sFlt-1 (soluble fms-like tyrosine kinase-1)/PlGF (placental growth factor) ratio. Chemerin release was studied in perfused placentas and placental explants with or without the statins pravastatin and fluvastatin. We also addressed statin placental passage and the effects of chemerin in chorionic plate arteries. RESULTS Serum chemerin was elevated in women with preeclampsia, and its addition to a predictive model yielded significant effects on top of the sFlt-1/PlGF ratio to predict preeclampsia and its fetal complications. Perfused placentas and explants of preeclamptic women released more chemerin and sFlt-1 and less PlGF than those of healthy pregnant women. Statins reversed this. Both statins entered the fetal compartment, and the fetal/maternal concentration ratio of pravastatin was twice that of fluvastatin. Chemerin constricted plate arteries, and this was blocked by a chemerin receptor antagonist and pravastatin. Chemerin did not potentiate endothelin-1 in chorionic plate arteries. In explants, statins upregulated low-density lipoprotein receptor expression, which relies on the same transcription factor as chemerin, and NO release. CONCLUSIONS Chemerin is a biomarker for preeclampsia, and statins both prevent its placental upregulation and effects, in an NO and low-density lipoprotein receptor-dependent manner. Combined with their capacity to improve the sFlt-1/PlGF ratio, this offers an attractive mechanism by which statins may prevent or treat preeclampsia.
Collapse
Affiliation(s)
- Lunbo Tan
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.T., A.C.M.K., E.O.C.-L., M.B., R.I.N., A.H.J.D., K.V.), Erasmus MC, Rotterdam, the Netherlands
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, China (L.T., X.L.)
| | - Ans C.M. Kluivers
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.T., A.C.M.K., E.O.C.-L., M.B., R.I.N., A.H.J.D., K.V.), Erasmus MC, Rotterdam, the Netherlands
- Department of Obstetrics and Gynecology (A.C.M.K., R.I.N., S.S.), Erasmus MC, Rotterdam, the Netherlands
| | - Edwyn O. Cruz-López
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.T., A.C.M.K., E.O.C.-L., M.B., R.I.N., A.H.J.D., K.V.), Erasmus MC, Rotterdam, the Netherlands
| | - Michelle Broekhuizen
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.T., A.C.M.K., E.O.C.-L., M.B., R.I.N., A.H.J.D., K.V.), Erasmus MC, Rotterdam, the Netherlands
- Division of Neonatology, Department of Neonatal and Pediatric Intensive Care (M.B.), Erasmus MC, Rotterdam, the Netherlands
| | - Zhongli Chen
- Department of Internal Medicine, Academic Center for Thyroid Diseases (Z.C.), Erasmus MC, Rotterdam, the Netherlands
| | - Rugina I. Neuman
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.T., A.C.M.K., E.O.C.-L., M.B., R.I.N., A.H.J.D., K.V.), Erasmus MC, Rotterdam, the Netherlands
- Department of Obstetrics and Gynecology (A.C.M.K., R.I.N., S.S.), Erasmus MC, Rotterdam, the Netherlands
| | - Sam Schoenmakers
- Department of Obstetrics and Gynecology (A.C.M.K., R.I.N., S.S.), Erasmus MC, Rotterdam, the Netherlands
| | - Liesbeth Ruijgrok
- Department of Hospital Pharmacy (L.R., D.v.d.V., B.C.M.d.W.), Erasmus MC, Rotterdam, the Netherlands
| | - Daan van de Velde
- Department of Hospital Pharmacy (L.R., D.v.d.V., B.C.M.d.W.), Erasmus MC, Rotterdam, the Netherlands
| | - Brenda C.M. de Winter
- Department of Hospital Pharmacy (L.R., D.v.d.V., B.C.M.d.W.), Erasmus MC, Rotterdam, the Netherlands
| | - Antoon J. van den Bogaerdt
- Heart Valve Department, Euro Tissue Bank-Bio Implant Services LIFE (ETB-BISLIFE), Beverwijk, the Netherlands (A.J.v.d.B.)
| | - Xifeng Lu
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, China (L.T., X.L.)
| | - A.H. Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.T., A.C.M.K., E.O.C.-L., M.B., R.I.N., A.H.J.D., K.V.), Erasmus MC, Rotterdam, the Netherlands
| | - Koen Verdonk
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (L.T., A.C.M.K., E.O.C.-L., M.B., R.I.N., A.H.J.D., K.V.), Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
125
|
Cooke WR, Jiang P, Ji L, Bai J, Jones GD, Lo YMD, Redman C, Vatish M. Differential 5'-tRNA Fragment Expression in Circulating Preeclampsia Syncytiotrophoblast Vesicles Drives Macrophage Inflammation. Hypertension 2024; 81:876-886. [PMID: 38362745 PMCID: PMC10956686 DOI: 10.1161/hypertensionaha.123.22292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/22/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND The relationship between placental pathology and the maternal syndrome of preeclampsia is incompletely characterized. Mismatch between placental nutrient supply and fetal demands induces stress in the syncytiotrophoblast, the layer of placenta in direct contact with maternal blood. Such stress alters the content and increases the release of syncytiotrophoblast extracellular vesicles (STB-EVs) into the maternal circulation. We have previously shown 5'-tRNA fragments (5'-tRFs) constitute the majority of small RNA in STB-EVs in healthy pregnancy. 5'-tRFs are produced in response to stress. We hypothesized STB-EV 5'-tRF release might change in preeclampsia. METHODS We perfused placentas from 8 women with early-onset preeclampsia and 6 controls, comparing small RNA expression in STB-EVs. We used membrane-affinity columns to isolate maternal plasma vesicles and investigate placental 5'-tRFs in vivo. We quantified 5'-tRFs from circulating STB-EVs using a placental alkaline phosphatase immunoassay. 5'-tRFs and scrambled RNA controls were added to monocyte, macrophage and endothelial cells in culture to investigate transcriptional responses. RESULTS 5'-tRFs constitute the majority of small RNA in STB-EVs from both preeclampsia and normal pregnancies. More than 900 small RNA fragments are differentially expressed in preeclampsia STB-EVs. Preeclampsia-dysregulated 5'-tRFs are detectable in maternal plasma, where we identified a placentally derived load. 5'-tRF-Glu-CTC, the most abundant preeclampsia-upregulated 5'-tRF in perfusion STB-EVs, is also increased in preeclampsia STB-EVs from maternal plasma. 5'-tRF-Glu-CTC induced inflammation in macrophages but not monocytes. The conditioned media from 5'-tRF-Glu-CTC-activated macrophages reduced eNOS (endothelial NO synthase) expression in endothelial cells. CONCLUSIONS Increased release of syncytiotrophoblast-derived vesicle-bound 5'-tRF-Glu-CTC contributes to preeclampsia pathophysiology.
Collapse
Affiliation(s)
- William R Cooke
- Nuffield Department of Women's and Reproductive Health, University of Oxford, United Kingdom (W.R.C., G.D.J., C.R., M.V.)
| | - Peiyong Jiang
- Centre for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, China (P.J., L.J., J.B., Y.M.D.L.)
- Department of Chemical Pathology, Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, China (P.J., L.J., J.B., Y.M.D.L.)
| | - Lu Ji
- Centre for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, China (P.J., L.J., J.B., Y.M.D.L.)
- Department of Chemical Pathology, Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, China (P.J., L.J., J.B., Y.M.D.L.)
| | - Jinyue Bai
- Centre for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, China (P.J., L.J., J.B., Y.M.D.L.)
- Department of Chemical Pathology, Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, China (P.J., L.J., J.B., Y.M.D.L.)
| | - Gabriel Davis Jones
- Nuffield Department of Women's and Reproductive Health, University of Oxford, United Kingdom (W.R.C., G.D.J., C.R., M.V.)
| | - Y M Dennis Lo
- Centre for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, China (P.J., L.J., J.B., Y.M.D.L.)
- Department of Chemical Pathology, Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, China (P.J., L.J., J.B., Y.M.D.L.)
| | - Christopher Redman
- Nuffield Department of Women's and Reproductive Health, University of Oxford, United Kingdom (W.R.C., G.D.J., C.R., M.V.)
| | - Manu Vatish
- Nuffield Department of Women's and Reproductive Health, University of Oxford, United Kingdom (W.R.C., G.D.J., C.R., M.V.)
| |
Collapse
|
126
|
Salazar MR. Placental hypoxia, high nighttime blood pressure, and maternal health. Hypertens Res 2024; 47:972-974. [PMID: 38182907 DOI: 10.1038/s41440-023-01567-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 01/07/2024]
Affiliation(s)
- Martin R Salazar
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Provincia de Buenos Aires, Argentina.
| |
Collapse
|
127
|
Zhu M, Liu J, Cao J, Ni Y, Chang M, Chen R, Su Z, Yu W, Ye H. Validation of a new kit for preeclampsia screening: A comprehensive analysis. Heliyon 2024; 10:e28080. [PMID: 38533029 PMCID: PMC10963371 DOI: 10.1016/j.heliyon.2024.e28080] [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: 10/04/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Objectives Preeclampsia is a common pregnancy complication that significantly contributes to maternal mortality, perinatal mortality, and preterm delivery. The sFlt-1/PlGF (fms-like tyrosine kinase-1/placental growth factor) ratio has demonstrated robust diagnostic value for preeclampsia. This study assessed the analytical performance and diagnostic accuracy of a novel quantitative determination kit for sFlt-1 and PlGF for the diagnosis of preeclampsia. Methods The detection performance of the test kit was validated using the Center for Medical Device Evaluation (CMDE) and Clinical and Laboratory Standards Institute (CLSI) documents. The test results were compared to those of the Elecsys immunoassay (Roche Diagnostics). Independent discovery and validation sets were used to analyze the diagnostic efficacy of the preeclampsia kit. The area under the curve (AUC) for preeclampsia at different gestational ages was calculated. Results Correlation analysis between the test and Roche kits revealed a strong concordance (sFlt-1: r = 0.9966, P < 0.0001; PlGF: r = 0.9935, P < 0.0001). The AUCs for sFlt-1, PlGF, and the sFlt-1/PlGF ratio in diagnosing preeclampsia were 0.749, 0.795, and 0.834, respectively, in the discovery set and 0.729, 0.811, and 0.831, respectively, in the validation set. The corresponding results from the Roche kit were 0.741, 0.795, and 0.829, respectively, and 0.761, 0.864, and 0.844, respectively. Conclusions Quantitative sFlt-1 and PlGF kits exhibited high levels of consistency with the Roche kits in terms of quantitative outcomes and diagnostic performance for preeclampsia.
Collapse
Affiliation(s)
- Min Zhu
- Department of Laboratory Medicine, Fujian Key Clinical Specialty of Laboratory Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Jumei Liu
- Department of Laboratory Medicine, Fujian Key Clinical Specialty of Laboratory Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Jiali Cao
- Department of Laboratory Medicine, Fujian Key Clinical Specialty of Laboratory Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Yan Ni
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Mengqi Chang
- School of Public Health, Xiamen University, Xiamen, China
| | - Ruitong Chen
- School of Public Health, Xiamen University, Xiamen, China
| | - Zhiying Su
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Weiwei Yu
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Huiming Ye
- Department of Laboratory Medicine, Fujian Key Clinical Specialty of Laboratory Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
128
|
Giardini V, Santagati AAF, Marelli E, Casati M, Cantarutti A, Vergani P. Predicting Time to Delivery in Hypertensive Disorders: Assessing PlGF and sFlt-1 with the Novel Parameter 'Mtp-Multiples of a Normal Term Placenta'. J Clin Med 2024; 13:1899. [PMID: 38610664 PMCID: PMC11012921 DOI: 10.3390/jcm13071899] [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: 03/03/2024] [Revised: 03/16/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Background: Imbalanced angiogenesis is characteristic of normal placental maturation but it also signals placental dysfunction, underlying hypertensive disorders during pregnancy. This study aimed to investigate the relationship between angiogenic placental aging, measured by markers placental growth factor (PlGF) and soluble fms-like tyrosine kinase-1 (sFlt-1) using the new index "Multiples of a normal term placenta" (Mtp) and the duration of pregnancy. Methods: A retrospective observational study was conducted, including singleton pregnancies diagnosed or suspected of hypertensive disorders after the 20th gestational week. Mtp measures how far a single dosage of angiogenic marker deviates from the expected value in an uncomplicated full-term pregnancy (Mpt = sFlt-1/sFlt-1 reference value or PIGF/PIGF reference value). We considered the 90th, 95th, and 97.5th centiles for sFlt-1 and the 2.5th, 5th, and 10th centiles for PlGF as references. Results: The categories with longer time to delivery, regardless of gestational age, were: Mtp PlGF 10th c ≥ 2, ≥3 and Mtp sFlt-1 90th c ≤ 0.5 (median days of 9, 11, 15 days, respectively). These two categories Mtp sFlt-1 90th c ≥ 3 and Mtp sFlt-1 97.5th c ≥ 2 allow the identification of women at risk for imminent delivery within 1 day. Women who were deemed at low/medium risk based on the sFlt-1/PIGF ratio appeared to be at high risk when considering the individual values of sFlt-1 and/or PIGF. Conclusions: This new Mtp index for sFlt-1 and PlGF could be employed to assess the degree of placental aging in women with hypertensive disorders. It represents a valid tool for evaluating the risk of imminent birth, irrespective of gestational age, surpassing the current stratification based on the sFlt-1/PIGF ratio.
Collapse
Affiliation(s)
- Valentina Giardini
- Department of Obstetrics, IRCCS San Gerardo dei Tintori Foundation, University of Milano-Bicocca, 20900 Monza, Italy
| | | | - Elisabetta Marelli
- Department of Obstetrics, MBBM Foundation Onlus, University of Milano-Bicocca, 20900 Monza, Italy
| | - Marco Casati
- Laboratory Medicine, IRCCS San Gerardo dei Tintori Foundation, 20900 Monza, Italy;
| | - Anna Cantarutti
- Division of Biostatistics, Epidemiology and Public Health, Department of Statistics and Quantitative Methods, University of Milano-Bicocca, 20126 Milano, Italy
| | - Patrizia Vergani
- Department of Obstetrics, MBBM Foundation Onlus, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
129
|
Parker J, O’Brien CL, Yeoh C, Gersh FL, Brennecke S. Reducing the Risk of Pre-Eclampsia in Women with Polycystic Ovary Syndrome Using a Combination of Pregnancy Screening, Lifestyle, and Medical Management Strategies. J Clin Med 2024; 13:1774. [PMID: 38541997 PMCID: PMC10971491 DOI: 10.3390/jcm13061774] [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: 02/12/2024] [Revised: 03/12/2024] [Accepted: 03/17/2024] [Indexed: 05/04/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is a multisystem disorder that presents with a variety of phenotypes involving metabolic, endocrine, reproductive, and psychological symptoms and signs. Women with PCOS are at increased risk of pregnancy complications including implantation failure, miscarriage, gestational diabetes, fetal growth restriction, preterm labor, and pre-eclampsia (PE). This may be attributed to the presence of specific susceptibility features associated with PCOS before and during pregnancy, such as chronic systemic inflammation, insulin resistance (IR), and hyperandrogenism, all of which have been associated with an increased risk of pregnancy complications. Many of the features of PCOS are reversible following lifestyle interventions such as diet and exercise, and pregnant women following a healthy lifestyle have been found to have a lower risk of complications, including PE. This narrative synthesis summarizes the evidence investigating the risk of PE and the role of nutritional factors in women with PCOS. The findings suggest that the beneficial aspects of lifestyle management of PCOS, as recommended in the evidence-based international guidelines, extend to improved pregnancy outcomes. Identifying high-risk women with PCOS will allow targeted interventions, early-pregnancy screening, and increased surveillance for PE. Women with PCOS should be included in risk assessment algorithms for PE.
Collapse
Affiliation(s)
- Jim Parker
- School of Medicine, University of Wollongong, Wollongong 2522, Australia
| | - Claire Louise O’Brien
- Faculty of Science and Technology, University of Canberra, Canberra 2617, Australia;
| | - Christabelle Yeoh
- Next Practice Genbiome, 2/2 New McLean Street, Edgecliff 2027, Australia;
| | - Felice L. Gersh
- College of Medicine, University of Arizona, Tucson, AZ 85004, USA;
| | - Shaun Brennecke
- Department of Maternal-Fetal Medicine, Pregnancy Research Centre, The Royal Women’s Hospital, Melbourne 3052, Australia;
- Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne 3052, Australia
| |
Collapse
|
130
|
Xia Y, Wang Y, Yuan S, Hu J, Zhang L, Xie J, Zhao Y, Hao J, Ren Y, Wu S. Development and validation of nomograms to predict clinical outcomes of preeclampsia. Front Endocrinol (Lausanne) 2024; 15:1292458. [PMID: 38549768 PMCID: PMC10972945 DOI: 10.3389/fendo.2024.1292458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/14/2024] [Indexed: 04/02/2024] Open
Abstract
Background Preeclampsia (PE) is one of the most severe pregnancy-related diseases; however, there is still a lack of reliable biomarkers. In this study, we aimed to develop models for predicting early-onset PE, severe PE, and the gestation duration of patients with PE. Methods Eligible patients with PE were enrolled and divided into a training (n = 253) and a validation (n = 108) cohort. Multivariate logistic and Cox models were used to identify factors associated with early-onset PE, severe PE, and the gestation duration of patients with PE. Based on significant factors, nomograms were developed and evaluated using the area under the curve (AUC) and a calibration curve. Results In the training cohort, multiple gravidity experience (p = 0.005), lower albumin (ALB; p < 0.001), and higher lactate dehydrogenase (LDH; p < 0.001) were significantly associated with early-onset PE. Abortion history (p = 0.017), prolonged thrombin time (TT; p < 0.001), and higher aspartate aminotransferase (p = 0.002) and LDH (p = 0.003) were significantly associated with severe PE. Abortion history (p < 0.001), gemellary pregnancy (p < 0.001), prolonged TT (p < 0.001), higher mean platelet volume (p = 0.014) and LDH (p < 0.001), and lower ALB (p < 0.001) were significantly associated with shorter gestation duration. Three nomograms were developed and validated to predict the probability of early-onset PE, severe PE, and delivery time for each patient with PE. The AUC showed good predictive performance, and the calibration curve and decision curve analysis demonstrated clinical practicability. Conclusion Based on the clinical features and peripheral blood laboratory indicators, we identified significant factors and developed models to predict early-onset PE, severe PE, and the gestation duration of pregnant women with PE, which could help clinicians assess the clinical outcomes early and design appropriate strategies for patients.
Collapse
Affiliation(s)
- Yan Xia
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Yao Wang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Shijin Yuan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaming Hu
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Lu Zhang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Jiamin Xie
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Yang Zhao
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Jiahui Hao
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yanwei Ren
- Department of Gynaecology and Obstetrics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shengjun Wu
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| |
Collapse
|
131
|
Nüsken E, Appel S, Saschin L, Kuiper-Makris C, Oberholz L, Schömig C, Tauscher A, Dötsch J, Kribs A, Alejandre Alcazar MA, Nüsken KD. Intrauterine Growth Restriction: Need to Improve Diagnostic Accuracy and Evidence for a Key Role of Oxidative Stress in Neonatal and Long-Term Sequelae. Cells 2024; 13:501. [PMID: 38534344 PMCID: PMC10969486 DOI: 10.3390/cells13060501] [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: 12/29/2023] [Revised: 03/01/2024] [Accepted: 03/11/2024] [Indexed: 03/28/2024] Open
Abstract
Intrauterine growth restriction (IUGR) and being small for gestational age (SGA) are two distinct conditions with different implications for short- and long-term child development. SGA is present if the estimated fetal or birth weight is below the tenth percentile. IUGR can be identified by additional abnormalities (pathological Doppler sonography, oligohydramnion, lack of growth in the interval, estimated weight below the third percentile) and can also be present in fetuses and neonates with weights above the tenth percentile. There is a need to differentiate between IUGR and SGA whenever possible, as IUGR in particular is associated with greater perinatal morbidity, prematurity and mortality, as well as an increased risk for diseases in later life. Recognizing fetuses and newborns being "at risk" in order to monitor them accordingly and deliver them in good time, as well as to provide adequate follow up care to ameliorate adverse sequelae is still challenging. This review article discusses approaches to differentiate IUGR from SGA and further increase diagnostic accuracy. Since adverse prenatal influences increase but individually optimized further child development decreases the risk of later diseases, we also discuss the need for interdisciplinary follow-up strategies during childhood. Moreover, we present current concepts of pathophysiology, with a focus on oxidative stress and consecutive inflammatory and metabolic changes as key molecular mechanisms of adverse sequelae, and look at future scientific opportunities and challenges. Most importantly, awareness needs to be raised that pre- and postnatal care of IUGR neonates should be regarded as a continuum.
Collapse
Affiliation(s)
- Eva Nüsken
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany; (E.N.)
| | - Sarah Appel
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany; (E.N.)
| | - Leon Saschin
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany; (E.N.)
| | - Celien Kuiper-Makris
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany; (E.N.)
| | - Laura Oberholz
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany; (E.N.)
| | - Charlotte Schömig
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany; (E.N.)
| | - Anne Tauscher
- Department of Obstetrics and Gynecology, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Jörg Dötsch
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany; (E.N.)
| | - Angela Kribs
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany; (E.N.)
| | - Miguel A. Alejandre Alcazar
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany; (E.N.)
- Institute for Lung Health (ILH), University of Giessen and Marburg Lung Center (UGMLC) and Cardiopulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), 35392 Giessen, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Kai-Dietrich Nüsken
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany; (E.N.)
| |
Collapse
|
132
|
Wang C, Ju H, Zhou L, Zhu Y, Wu L, Deng X, Jiang L, Sun L, Xu Y. TET3-mediated novel regulatory mechanism affecting trophoblast invasion and migration: Implications for preeclampsia development. Placenta 2024; 147:31-41. [PMID: 38295560 DOI: 10.1016/j.placenta.2024.01.010] [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: 10/11/2023] [Revised: 12/28/2023] [Accepted: 01/21/2024] [Indexed: 02/02/2024]
Abstract
INTRODUCTION Aberrant expression of genes has been demonstrated to be related to the abnormal function of trophoblasts and lead to the occurrence and progression of Preeclampsia (PE). However, the underlying mechanism of PE has not been elucidated. METHODS We performed PCR analysis to investigate TET3 expression in PE placental tissues. Cell assays were performed in HTR-8/SVneo and JAR. Cell invasion and migration events were investigated by transwell assays in vitro. ChIP-PCR and Targeted bisulfite sequencing were conducted to detect the demethylation of related CpG sites in the KLF13 promoter after inhibition of TET3. In conjunction with bioinformatics analysis, luciferase reporter assays were performed to elucidate the mechanism by which miR-544 binds to TET3/KLF13 mRNA. RESULTS In this study, we identified genes associated with human extravillous trophoblasts by conducting sc-seq analysis from the GEO. Then, we measured the expression of TET3 in a larger clinical sample. The results showed that TET3, a DNA demethylase, was found to be expressed at much higher levels in the preeclamptic placenta compared to the control. Then, the inhibition of TET3 significantly promoted trophoblast cell migration and invasion. Conversely, TET3 overexpression suppressed cell migration and invasion in vitro. Further RNA sequencing and mechanism analysis indicated that the inhibition of TET3 suppressed the activation of KLF13 by reducing the demethylation of related CpG sites in the KLF13 promoter, thereby transcriptionally inactivating KLF13 expression. Moreover, luciferase reporter assay indicate that TET3 and KLF13 were direct targets of miR-544. DISCUSSION This study uncovers a TET3-mediated regulatory mechanism in PE progression and suggests that targeting the placental miR-544-TET3-KLF13-axis might provide new diagnostic and therapeutic strategies for PE.
Collapse
Affiliation(s)
- Cong Wang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Huihui Ju
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China; Department of Obstetrics and Gynecology, Changzhou Maternal and Child Health Care Hospital Changzhou Medical Center of Nanjing Medical University, Changzhou, 213000, Jiangsu Province, China
| | - Lihong Zhou
- Department of Cardiovascular Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Yuanyuan Zhu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Liuxin Wu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Xiaokang Deng
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Lingling Jiang
- Department of Obstetrics and Gynecology, Affiliated Hospital 2 of Nantong University, Nantong, 226001, Jiangsu Province, China.
| | - Lizhou Sun
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.
| | - Yetao Xu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
133
|
Paquin A, Wei J. Prediction of Preeclampsia: Time for the Cardiovascular Community to Be Involved. Can J Cardiol 2024; 40:431-433. [PMID: 38016542 DOI: 10.1016/j.cjca.2023.11.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 11/30/2023] Open
Affiliation(s)
- Amélie Paquin
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada; Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada; Barbra Streisand Women's Heart Center, Cedars-Sinai Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| | - Janet Wei
- Barbra Streisand Women's Heart Center, Cedars-Sinai Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
134
|
Daskalopoulou SS, Labos C, Kuate Defo A, Cooke AB, Kalra B, Kumar A, Mantzoros CS. Analysis of Predictive Information From Biomarkers Added to Clinical Models of Preeclampsia: Consideration of PAPP-A2, Activin A, and sFlt-1:PlGF Ratio. Can J Cardiol 2024; 40:422-430. [PMID: 38787345 DOI: 10.1016/j.cjca.2023.10.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Preeclampsia remains a major cause of maternal and fetal adverse outcomes in pregnancy; however, accurate and universally acceptable predictive tools remain elusive. We investigated whether a panel of biomarkers could improve risk prediction for preeclampsia when measured at various pregnancy time points. METHODS In this prospective cohort study, 192 women with first-trimester high-risk singleton pregnancies were consecutively recruited from tertiary obstetrics clinics in Montréal, Canada. Clinical information (height, pre-pregnancy weight, personal and family medical history, medication use) was collected at baseline. Blood pressure was measured and blood samples collected at each trimester to quantify soluble Fms-like tyrosine kinase 1 (sFlt-1), placental growth factor (PlGF), pregnancy-associated plasma protein A2 (PAPP-A2), PAPP-A, activin A, inhibin A, follistatin, and glycosylated fibronectin. A random-effects hierarchic logistic regression model was used to relate change in biomarker levels to incidence of preeclampsia. RESULTS When added to a clinical model composed of maternal age, pre-pregnancy body mass index, race, and mean arterial pressure, a positive third-trimester result for both PAPP-A2 and activin A had a better positive predictive value than the sFlt-1:PlGF ratio added to the clinical model (91.67% [95% confidence interval (CI) 78.57%-100%] vs 66.67% [57.14%-100%]), while maintaining a comparable high negative predictive value (97.69% [95% CI 95.34%-100%] vs 96.00% [92.19%-99.21%]). CONCLUSIONS Whereas the third-trimester sFlt-1:PlGF ratio can predict short-term absence of preeclampsia, PAPP-A2 and activin A had both high positive and negative predictive values and therefore could serve as biomarkers to predict the occurrence (and absence) of preeclampsia; these findings will be validated in future studies.
Collapse
Affiliation(s)
- Stella S Daskalopoulou
- Vascular Health Unit, Research Institute of the McGill University Health Centre, Department of Medicine, McGill University, Montréal, Québec, Canada; Division of Experimental Medicine, Department of Medicine, McGill University, Montréal, Québec, Canada; Division of Internal Medicine, Department of Medicine, McGill University Health Centre, McGill University, Montréal, Québec, Canada.
| | - Christopher Labos
- Vascular Health Unit, Research Institute of the McGill University Health Centre, Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Alvin Kuate Defo
- Vascular Health Unit, Research Institute of the McGill University Health Centre, Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Alexandra B Cooke
- Division of Experimental Medicine, Department of Medicine, McGill University, Montréal, Québec, Canada
| | | | | | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
135
|
Yang M, Bai Y, Li M, Lin X, Duan X, Zhang X. Predictive value of the soluble fms-like tyrosine kinase 1 to placental growth factor ratio for preeclampsia in twin pregnancies: a systematic review and meta-analysis. Am J Obstet Gynecol MFM 2024; 6:101290. [PMID: 38401234 DOI: 10.1016/j.ajogmf.2024.101290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/04/2024] [Accepted: 01/11/2024] [Indexed: 02/26/2024]
Abstract
OBJECTIVE In recent years, the ratio of soluble fms-like tyrosine kinase 1 to placental growth factor for use in predicting preeclampsia has been explored extensively. Despite extensive research, available data on its effectiveness in predicting preeclampsia in twin pregnancies are limited and conflicting. This meta-analysis aimed to assess the diagnostic accuracy of the soluble fms-like tyrosine kinase 1 to placental growth factor ratio in distinguishing cases with preeclampsia in twin pregnancies from healthy controls. DATA SOURCES Studies that evaluated the use of the soluble fms-like tyrosine kinase 1 to placental growth factor ratio in predicting preeclampsia were searched in PubMed, Embase, and Cochrane databases from inception to August 6, 2023, without language restriction. STUDY ELIGIBILITY CRITERIA The following population, exposure, comparators, outcomes, and study designs were included: women with twin pregnancies; an increased soluble fms-like tyrosine kinase 1 to placental growth factor ratio with preeclampsia as the outcome; women without preeclampsia; a 2 × 2 diagnostic table, diagnostic accuracy data, and the incidence of preeclampsia; and prospective cohort studies and observational comparative studies, respectively. STUDY APPRAISAL AND SYNTHESIS METHODS The quality of the included studies was evaluated. Key parameters, including the specificity, sensitivity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio, were calculated using the random- and fixed-effects models. In addition, the area under the receiver operating characteristic curve and the summary receiver operating characteristic curve were evaluated. RESULTS A total of 7 studies were included, including 442 women with twin pregnancies (115 patients with preeclampsia and 327 controls without preeclampsia). The results highlighted the promising effectiveness of the soluble fms-like tyrosine kinase 1 to placental growth factor ratio in predicting preeclampsia in twin pregnancies with a pooled specificity of 0.89 (95% confidence interval, 0.80-0.95), a sensitivity of 0.84 (95% confidence interval, 0.73-0.93), a positive likelihood ratio of 32.76 (95% confidence interval, 12.82-83.74), and a negative likelihood ratio of 0.03 (95% confidence interval, 0.01-0.08). The combined diagnostic odds ratio was 35.72 (95% confidence interval, 12.92-98.76), and the area under the receiver operating characteristic curve was 0.92. CONCLUSION These collective findings underscore the potential of the soluble fms-like tyrosine kinase 1 to placental growth factor ratio as an accurate marker for identifying preeclampsia among women with twin pregnancies.
Collapse
Affiliation(s)
- Meilin Yang
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China (Drs Yang, Lin, Duan, and Zhang); Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetric Diseases and Department of Obstetrics, Women and Children's Hospital, Xiamen University, Xiamen, China (Drs Yang, Lin, Duan, and Zhang)
| | - Yuci Bai
- Department of Obstetrics and Gynecology, Xiamen Chang Gung Hospital, Xiamen, Fujian, China (Drs Bai and Li)
| | - Min Li
- Department of Obstetrics and Gynecology, Xiamen Chang Gung Hospital, Xiamen, Fujian, China (Drs Bai and Li)
| | - Xueyan Lin
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China (Drs Yang, Lin, Duan, and Zhang); Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetric Diseases and Department of Obstetrics, Women and Children's Hospital, Xiamen University, Xiamen, China (Drs Yang, Lin, Duan, and Zhang)
| | - Xiaoyu Duan
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China (Drs Yang, Lin, Duan, and Zhang); Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetric Diseases and Department of Obstetrics, Women and Children's Hospital, Xiamen University, Xiamen, China (Drs Yang, Lin, Duan, and Zhang)
| | - Xueqin Zhang
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China (Drs Yang, Lin, Duan, and Zhang); Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetric Diseases and Department of Obstetrics, Women and Children's Hospital, Xiamen University, Xiamen, China (Drs Yang, Lin, Duan, and Zhang).
| |
Collapse
|
136
|
Ramírez Sanchez FA, Madrigal Aguilar D, Tufiño C, Castro García S, Bobadilla Lugo RA. Preeclampsia pravastatin early VS late treatment: Effects on oxidative stress and vascular reactivity. Pregnancy Hypertens 2024; 35:96-102. [PMID: 38306739 DOI: 10.1016/j.preghy.2024.01.131] [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: 05/02/2023] [Revised: 01/05/2024] [Accepted: 01/13/2024] [Indexed: 02/04/2024]
Abstract
Early diagnosis and efficient treatment of preeclampsia remains a medical challenge and etiological factors converge in a deficient placentation that triggers oxidative stress. There is evidence that statins show antioxidant effects that can improve endothelial function without adverse perinatal effects. We aimed to compare early vs. late pravastatin treatment on the oxidative stress and cardiovascular features of an experimental model of preeclampsia. Female Wistar rats were randomly divided into preeclampsia phenotype rats (PEP) developed by sub renal aortic coarctation (SRAC) and healthy pregnant rats (C). Each group received pravastatin (5 mg/Kg) p.o. either for one week before and during the first week or during the last two weeks of gestation. Blood pressure was determined using the plethysmographic method. Phenylephrine (Phe)-induced contractility was evaluated in isolated thoracic and abdominal aortic rings with or without endothelium. Blood samples were obtained to determine anion superoxide concentration as indicator of NADPH activity. Two-way ANOVA and Bonferroni post hoc tests were used to define statistical significance. Early or late pravastatin treatment decreased hypertension of PEP animals but did not change BP of the healthy pregnant group. Thoracic and abdominal aorta from PEP rats showed increased contractility that was reverted by pravastatin early treatment in endothelium intact rings. Pravastatin did not significantly change contractility neither in the thoracic nor in the abdominal aorta segments from healthy pregnant control rats (C), and decrease anion superoxide concentration by NADPH activity. We conclude pravastatin can improve both blood pressure and endothelium-dependent Phe-induced contractility in an experimental model of preeclampsia by reducing oxidative stress.
Collapse
Affiliation(s)
| | | | - Cecilia Tufiño
- Escuela Superior de Medicina Instituto Politécnico Nacional, Mexico City, Mexico
| | - Seidy Castro García
- Escuela Superior de Medicina Instituto Politécnico Nacional, Mexico City, Mexico
| | | |
Collapse
|
137
|
Ma'ayeh M, Cavus O, Hassen LJ, Johnson M, Summerfield T, Begom M, Cai A, Mehta L, Rood K, Bradley EA. Study of heart function in PRE-Eclampsia during and after PreGnancy (SHePREG): The pilot cohort. Am Heart J 2024; 269:45-55. [PMID: 38103586 PMCID: PMC10922975 DOI: 10.1016/j.ahj.2023.12.003] [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: 08/18/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND Pre-eclampsia with severe features (severe PreE) is associated with heart dysfunction, yet the impact beyond pregnancy, including its association with cardiomyopathic genetic polymorphisms, remains poorly understood. OBJECTIVE We aimed to characterize the temporal impact of severe PreE on heart function through the 4th trimester in women with and without deleterious cardiomyopathic genetic variants. METHODS Pregnant women were enrolled to undergo transthoracic echocardiography (TTE) in late pregnancy and 3 months postpartum. In women with severe PreE a targeted approach to identify pathogenic cardiomyopathic genetic polymorphisms was undertaken, and heart function was compared in carriers and noncarriers. RESULTS Pregnant women (32 ± 4 years old, severe PreE = 14, control = 8) were enrolled between 2019 - 2021. Women with severe PreE displayed attenuated myocardial relaxation (mitral e' = 11.0 ± 2.2 vs 13.2 ± 2.3 cm/sec, P < .05) in late pregnancy, and on in-silico analysis, deleterious cardiomyopathic variants were found in 58%. At 103 ± 33 days postpartum, control women showed stability in myocardial relaxation (Mitral e' Entry: 13.2 ± 2.3 vs Postpartum: 13.9 ± 1.7cm/sec, P = .464), and genetic negative severe PreE women (G-) demonstrated recovery of diastolic function to control level (Mitral e' Entry: 11.0 ± 3.0 vs Postpartum 13.7 ± 2.8cm/sec, P < .001), unlike their genetic positive (G+) counterparts (Mitral e' Entry: 10.5 ± 1.7 vs Postpartum 10.8 ± 2.4cm/sec, P = .853). CONCLUSIONS Postpartum recovery of heart function after severe PreE is attenuated in women with deleterious cardiomyopathic genetic polymorphisms.
Collapse
Affiliation(s)
- Marwan Ma'ayeh
- Division of Maternal Fetal Medicine, Christiana Hospital, Department of Obstetrics and Gynecology, Newark, DE
| | - Omer Cavus
- Pennsylvania State University Hershey S. Milton Medical Center, Heart and Vascular Institute, Division of Cardiovascular Medicine, Hershey, PA
| | - Lauren J Hassen
- The Ohio State University, Department of Internal Medicine, Division of Cardiovascular Medicine, Columbus, OH
| | - Martin Johnson
- Pennsylvania State University College of Medicine, Hershey PA
| | - Taryn Summerfield
- The Ohio State University, Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Columbus, OH
| | - Mosammat Begom
- Pennsylvania State University Hershey S. Milton Medical Center, Heart and Vascular Institute, Division of Cardiovascular Medicine, Hershey, PA
| | - Amanda Cai
- Pennsylvania State University Hershey S. Milton Medical Center, Heart and Vascular Institute, Division of Cardiovascular Medicine, Hershey, PA
| | - Laxmi Mehta
- The Ohio State University, Department of Internal Medicine, Division of Cardiovascular Medicine, Columbus, OH
| | - Kara Rood
- The Ohio State University, Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Columbus, OH
| | - Elisa A Bradley
- Pennsylvania State University Hershey S. Milton Medical Center, Heart and Vascular Institute, Division of Cardiovascular Medicine, Hershey, PA; Pennsylvania State University College of Medicine, Hershey PA; Pennsylvania State University, College of Medicine, Department of Cellular and Molecular Physiology, Hershey, PA.
| |
Collapse
|
138
|
Mecacci F, Romani E, Clemenza S, Zullino S, Avagliano L, Petraglia F. Early Fetal Growth Restriction with or Without Hypertensive Disorders: a Clinical Overview. Reprod Sci 2024; 31:591-602. [PMID: 37684516 DOI: 10.1007/s43032-023-01330-9] [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: 04/05/2023] [Accepted: 08/14/2023] [Indexed: 09/10/2023]
Abstract
Early onset fetal growth restriction (FGR) is one of the main adverse pregnancy conditions, often associated with poor neonatal outcomes. Frequently, early onset FGR is associated with early onset hypertensive disorders of pregnancy (HDP), and in particular preeclampsia (PE). However, to date, it is still an open question whether pregnancies complicated by early FGR plus HDP (FGR-HDP) and those complicated by early onset FGR without HDP (normotensive-FGR (n-FGR)) show different prenatal and postnatal outcomes and, consequently, should benefit from different management and long-term follow-up. Recent data support the hypothesis that the presence of PE may have an additional impact on maternal hemodynamic impairment and placental lesions, increasing the risk of poor neonatal outcomes in pregnancy affected by early onset FGR-HDP compared to pregnancy affected by early onset n-FGR. This review aims to elucidate this poor studied topic, comparing the clinical characteristics, perinatal outcomes, and potential long-term sequelae of early onset FGR-HDP and early onset n-FGR.
Collapse
Affiliation(s)
- Federico Mecacci
- Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and Gynecology, University of Florence, Florence, Italy
| | - Eleonora Romani
- Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and Gynecology, University of Florence, Florence, Italy
| | - Sara Clemenza
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Sara Zullino
- Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and Gynecology, University of Florence, Florence, Italy
| | | | - Felice Petraglia
- Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and Gynecology, University of Florence, Florence, Italy
| |
Collapse
|
139
|
Liu X, Xu J, Liu C, Li X. CercaTest Red, a novel urine-based point-of-care test for the detection of preeclampsia. Curr Med Res Opin 2024; 40:395-401. [PMID: 38321953 DOI: 10.1080/03007995.2024.2314245] [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: 12/11/2023] [Accepted: 01/31/2024] [Indexed: 02/08/2024]
Abstract
OBJECTIVES Preeclampsia (PE) poses a serious threat to the health of the pregnant woman and her developing fetus due to the difficulty in diagnosing the condition. The disease can develop and worsen suddenly without noticeable signs and symptoms. Thus, there is an urgent need for a simple Point of Care Test (POCT) that improves accessibility to testing and can be used as an aid in the diagnosis of PE. CercaTest Red is a noninvasive detection kit for impending preeclampsia using urine from pregnant women. This is especially pertinent for women who have limited access to secondary/tertiary healthcare as those in remote settings, low-income countries or simply lack of out of hours laboratory services. METHODS The kit employs an absorptive column that separates Congo red dye bound to urinary misfolded protein from pregnant women and unbound dye. When a solution of Congo red dye pre-mixed with urine is loaded onto the absorptive matrix in a detection cuvette, the presence (positive) or absence (negative) of misfolded proteins can be determined based on the color of eluate collected in the lower section of the cuvette. 190 and 937 pregnant women who were >18 years old at the gestational age of ≥20 weeks were enrolled for the feasibility and validation cohort, respectively. The consistency between CercaTest Red and clinical diagnosis of PE according to the American College of Obstetricians and Gynecologist (ACOG) Guidelines was analyzed using the kappa statistic. RESULTS The POCT has a limit of detection (LoD) of human urinary misfolded proteins equivalent to 0.45 μM of denatured human serum albumin, with high reproducibility and stability. An accuracy of 96.84% for diagnosis of preeclampsia with a Kappa statistic of 0.746 (p < 0.001) was validated in a cohort of 937 subjects. CONCLUSION This test is easy to use, cost-effective and portable with short turnaround time and no laboratory instrument requirement. In the future, the test may have the potential to become quantitative using spectroscopy (Chinese Clinical Trial Registry No. ChiCTR1800017692).
Collapse
Affiliation(s)
- Xuemin Liu
- Department of Gynaecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jun Xu
- Shuwen Biotech Co., Ltd, Hangzhou, Zhejiang, China
| | - Chao Liu
- Shuwen Biotech Co., Ltd, Hangzhou, Zhejiang, China
| | - Xingmin Li
- Shuwen Biotech Co., Ltd, Hangzhou, Zhejiang, China
- Cerca Biotech GmbH, Berlin, Germany
| |
Collapse
|
140
|
Udeh PI, Olumodeji AM, Kuye-Kuku TO, Orekoya OO, Ayanbode O, Fabamwo AO. Evaluating mean platelet volume and platelet distribution width as predictors of early-onset pre-eclampsia: a prospective cohort study. Matern Health Neonatol Perinatol 2024; 10:5. [PMID: 38424566 PMCID: PMC10905831 DOI: 10.1186/s40748-024-00174-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/17/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Platelets are pivotal players in the pathophysiology of pre-eclampsia, with observed lower counts in affected individuals compared to normotensive counterparts. Despite advancements, the elusive cause of pre-eclampsia persists, motivating intense global efforts to identify reliable predictors. The currently recommended predictors of pre-eclampsia are not readily available in many resource-limited regions like Nigeria. This cohort study explores the potential of mean platelet volume (MPV) and platelet distribution width (PDW) as predictive markers of early-onset pre-eclampsia. Both platelet indices are components of the full blood count, a widely available routine test in pregnancy. METHODS In this prospective cohort study, 648 healthy pregnant women attending antenatal care at Lagos State University Teaching Hospital and General Hospital Ifako-Ijaiye, Lagos, were recruited between 14-18weeks gestational age. Platelet count (PC), MPV and PDW were measured from their venous blood at recruitment. Participants were monitored until 34weeks of gestation, focusing on the occurrence of early-onset preeclampsia as the outcome of interest. Individuals with chronic medical conditions were excluded from the study. Data analysis involved t-test, Chi-Square and Mann-Whitney U tests, with statistical significance set at a confidence level of 95% and p < 0.05. Sensitivity, specificity, and predictive values were determined using receiver operating characteristics (ROC) curves. RESULTS The incidence of early-onset pre-eclampsia in the study was 5.9%. Women who later developed pre-eclampsia had higher median MPV and PDW at 14-18weeks (10.8 fl. and 24.8 fl.) compared to normotensive women (8.1 fl. and 13.3 fl.)(p < 0.001). The median PC was lower in pre-eclamptics (190 × 103/µl) compared to normotensives(264 × 103/µl)(p < 0.001). Using Youden's test, cut-off values identified: PC < 211.5 × 103/µl, MPV > 9.4 fl., and PDW > 21.3 fl., predicted early-onset pre-eclampsia with 96.6% sensitivity and 65.6% specificity for PC; 79.3% sensitivity and 97.7% specificity for PDW; and 82.8% sensitivity and 96.1% specificity for MPV. Cut-offs of PC < 185 × 103/µl, MPV > 10.7 fl., and PDW > 28.3 fl., predicted severe early-onset pre-eclampsia with 100.0% sensitivity and 90.9% specificity for PC, 100.0% sensitivity and 99.4% specificity for MPV, and 100.0% sensitivity and 99.8% specificity for PDW, with corresponding area under the ROC curves of 0.983, 0.996, and 0.998, respectively. CONCLUSION The evaluation of MPV and PDW between 14 and 18 weeks of gestation appears to be a reliable predictor of severe early-onset pre-eclampsia.
Collapse
Affiliation(s)
- Patience Ijeoma Udeh
- Department of Obstetrics and Gynaecology, Lagos State University Teaching Hospital, Lagos, Nigeria
| | - Ayokunle Moses Olumodeji
- Department of Obstetrics and Gynaecology, Lagos State University Teaching Hospital, Lagos, Nigeria.
| | - Taiwo Olufunmilayo Kuye-Kuku
- Department of Obstetrics and Gynaecology, Lagos State University Teaching Hospital, Lagos, Nigeria
- Department of Obstetrics and Gynaecology, Lagos State University College of Medicine, Lagos, Nigeria
| | | | - Olufemi Ayanbode
- Department of Obstetrics and Gynaecology, Lagos State University Teaching Hospital, Lagos, Nigeria
| | - Adetokunbo Olusegun Fabamwo
- Department of Obstetrics and Gynaecology, Lagos State University Teaching Hospital, Lagos, Nigeria
- Department of Obstetrics and Gynaecology, Lagos State University College of Medicine, Lagos, Nigeria
| |
Collapse
|
141
|
Montenegro-Martínez J, Camacho-Carrasco A, Nuñez-Jurado D, Beltrán-Romero LM, Fatela-Cantillo D. Longitudinal changes of angiogenic factors as a potential predictive tool in women with suspected preeclampsia. Pregnancy Hypertens 2024; 35:66-72. [PMID: 38245918 DOI: 10.1016/j.preghy.2024.01.130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/13/2024] [Indexed: 01/23/2024]
Abstract
OBJECTIVES To investigate whether longitudinal changes of angiogenic factors (AF) sFlt-1, PlGF, and the sFlt-1/PlGF ratio, measured following identification of symptoms of preeclampsia (PE), could provide complementary information to the isolated measurements used in current clinical practice. STUDY DESIGN Retrospective observational study. Sixty women with suspected PE and two AF results measured before gestational week (GW) 34 were included. Daily variation (DV) of AF was calculated from delta values and days elapsed between measurements. Through ROC analysis, the predictive performance of DV for PE-related events was estimated. Kaplan-Meier survival curves resulting from applying cutoff values were assessed. RESULTS The sFlt-1, PlGF, and sFlt-1/PlGF ratio baseline levels showed significant differences between women without PE and women who developed early-onset PE (P < 0.001). DV of sFlt-1 and sFlt-1/PlGF ratio increased according to the severity of PE, showing significant differences in both pairs of groups compared (p < 0.001), so they were selected as potential predictors. Higher AUC values resulting from ROC analysis were 0.78 for early-onset PE, 0.88 for early-onset severe PE, 0.79 for occurrence of adverse maternal outcomes, and 0.89 for delivery before 37 GW, with sensitivity and specificity values higher than 0.71 and 0.80, respectively. The Kaplan-Meier analysis yielded significantly different curves (log-rank < 0.05), with shorter time-to-delivery as DV increased. CONCLUSION Our results support the existence of a correlation between a progressive PlGF and sFlt-1 imbalance and a more aggressive clinical course of PE, detectable from the finding of PE symptoms. Its monitoring could be a useful predictive tool in women with suspected PE.
Collapse
Affiliation(s)
- Jorge Montenegro-Martínez
- Department of Clinical Biochemistry, Virgen del Rocío University Hospital, Laboratory Building, 4th Floor, Manuel Siurot Avenue, 41013 Seville, Spain.
| | - Ana Camacho-Carrasco
- Internal Medicine, Virgen del Rocío University Hospital, General Hospital Building, 2th Floor, Manuel Siurot Avenue, 41013 Seville, Spain
| | - David Nuñez-Jurado
- Department of Clinical Biochemistry, Virgen del Rocío University Hospital, Laboratory Building, 4th Floor, Manuel Siurot Avenue, 41013 Seville, Spain
| | - Luis M Beltrán-Romero
- Internal Medicine, Virgen del Rocío University Hospital, General Hospital Building, 2th Floor, Manuel Siurot Avenue, 41013 Seville, Spain
| | - Daniel Fatela-Cantillo
- Department of Clinical Biochemistry, Virgen del Rocío University Hospital, Laboratory Building, 4th Floor, Manuel Siurot Avenue, 41013 Seville, Spain
| |
Collapse
|
142
|
Shinohara S, Yoshihara T, Mochizuki K, Yasuda G, Kasai M, Sunami R. Preeclampsia prediction model using demographic, clinical, and sonographic data in the second trimester of Japanese nulliparous women. J Obstet Gynaecol Res 2024; 50:395-402. [PMID: 38109933 DOI: 10.1111/jog.15861] [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: 08/02/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023]
Abstract
AIM This study aimed to clarify the factors influencing preeclampsia (PE) development in nulliparous Japanese women and to develop a PE prediction model using second trimester sonographic and clinical data readily available to obstetricians. METHODS This historical cohort study examined the obstetric records of nulliparous women who delivered at Yamanashi Prefectural Central Hospital from January 2019 to May 2023. A model was constructed to predict the PE development rate, with a focus on 796 nulliparous women. The assessed outcome was PE, excluding superimposed PE. Data on maternal age, assisted reproductive technology, mean arterial pressure, uterine artery notching, and umbilical artery resistance index were extracted. Multivariable logistic regression analysis was conducted on these five factors. RESULTS The incidence of PE was 4.3% (34/796). Multivariable analysis indicated significant odds ratios for the association of PE with mean arterial pressure (adjusted odds ratio: 1.06, 95% confidence interval: 1.03-1.10) and uterine artery notching (adjusted odds ratio: 6.28, 95% confidence interval: 2.82-14.0) in nulliparous women. The PE prediction formula was established as follows: Probability of PE development (%) = (odds/1 + odds) × 100, odds = ex and x = -11.3 + 0.039 × maternal age (years) + 0.91 × assisted reproductive technology + 0.061 × mean arterial pressure (mmHg) + 1.84 × uterine artery notching + 1.84 × umbilical artery resistance index. The sensitivity and specificity of this model were 58.8% and 84.5%, respectively (area under the curve: 0.79). CONCLUSIONS This study is the first to provide a prediction formula targeting the Japanese population. Our specialized model for nulliparous women could guide obstetricians to educate women regarding the precise prospect of PE development.
Collapse
Affiliation(s)
- Satoshi Shinohara
- Department of Obstetrics and Gynecology, Yamanashi Prefectural Central Hospital, Kofu, Yamanashi, Japan
| | - Tatsuya Yoshihara
- Department of Obstetrics and Gynecology, Yamanashi Prefectural Central Hospital, Kofu, Yamanashi, Japan
| | - Kana Mochizuki
- Department of Obstetrics and Gynecology, Yamanashi Prefectural Central Hospital, Kofu, Yamanashi, Japan
| | - Genki Yasuda
- Department of Obstetrics and Gynecology, Yamanashi Prefectural Central Hospital, Kofu, Yamanashi, Japan
| | - Mayuko Kasai
- Department of Obstetrics and Gynecology, Yamanashi Prefectural Central Hospital, Kofu, Yamanashi, Japan
| | - Rei Sunami
- Department of Obstetrics and Gynecology, Yamanashi Prefectural Central Hospital, Kofu, Yamanashi, Japan
| |
Collapse
|
143
|
Mapari SA, Shrivastava D, Bedi GN, Pradeep U. Unveiling the Hidden Culprit of Posterior Reversible Encephalopathy Syndrome Superimposing Postpartum Eclampsia: A Rare Case. Cureus 2024; 16:e57006. [PMID: 38681382 PMCID: PMC11046172 DOI: 10.7759/cureus.57006] [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: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 05/01/2024] Open
Abstract
The rare yet potentially fatal neurological complication known as posterior reversible encephalopathy syndrome (PRES) can manifest during pregnancy. Alongside symptoms such as headaches, nausea, visual disturbances, and altered mental status, patients often experience seizures or loss of consciousness. Imaging typically reveals vascular edema affecting the parietal and occipital lobes within the subcortical region. We present the case of a 24-year-old patient who developed postpartum eclampsia followed by PRES. MRI findings demonstrated hyperintensities in the posterior parietal, frontal, and occipital lobes bilaterally, confirming the diagnosis. Prompt administration of levetiracetam and labetalol led to the resolution of the patient's symptoms. Subsequently, we thoroughly searched online databases for peer-reviewed articles examining the etiology, clinical presentation, and treatment options for PRES. Our evaluation of the case findings alongside existing literature underscored the rarity of PRES occurring concurrently with postpartum eclampsia, highlighting the importance of timely identification and intervention in managing this condition. Further research is warranted to enhance our understanding of PRES in the context of pregnancy-related complications.
Collapse
Affiliation(s)
- Smruti A Mapari
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Deepti Shrivastava
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Gautam N Bedi
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Utkarsh Pradeep
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
144
|
Frimat M, Gnemmi V, Stichelbout M, Provôt F, Fakhouri F. Pregnancy as a susceptible state for thrombotic microangiopathies. Front Med (Lausanne) 2024; 11:1343060. [PMID: 38476448 PMCID: PMC10927739 DOI: 10.3389/fmed.2024.1343060] [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: 11/22/2023] [Accepted: 02/12/2024] [Indexed: 03/14/2024] Open
Abstract
Pregnancy and the postpartum period represent phases of heightened vulnerability to thrombotic microangiopathies (TMAs), as evidenced by distinct patterns of pregnancy-specific TMAs (e.g., preeclampsia, HELLP syndrome), as well as a higher incidence of nonspecific TMAs, such as thrombotic thrombocytopenic purpura or hemolytic uremic syndrome, during pregnancy. Significant strides have been taken in understanding the underlying mechanisms of these disorders in the past 40 years. This progress has involved the identification of pivotal factors contributing to TMAs, such as the complement system, ADAMTS13, and the soluble VEGF receptor Flt1. Regardless of the specific causal factor (which is not generally unique in relation to the usual multifactorial origin of TMAs), the endothelial cell stands as a central player in the pathophysiology of TMAs. Pregnancy has a major impact on the physiology of the endothelium. Besides to the development of placenta and its vascular consequences, pregnancy modifies the characteristics of the women's microvascular endothelium and tends to render it more prone to thrombosis. This review aims to delineate the distinct features of pregnancy-related TMAs and explore the contributing mechanisms that lead to this increased susceptibility, particularly influenced by the "gravid endothelium." Furthermore, we will discuss the potential contribution of histopathological studies in facilitating the etiological diagnosis of pregnancy-related TMAs.
Collapse
Affiliation(s)
- Marie Frimat
- CHU Lille, Nephrology Department, Univ. Lille, Lille, France
- Inserm, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | | | | | - François Provôt
- CHU Lille, Nephrology Department, Univ. Lille, Lille, France
| | - Fadi Fakhouri
- Service of Nephrology and Hypertension, CHUV and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
145
|
Cerdeira AS, Karumanchi SA. Serial placental growth factor-based testing in pre-eclampsia. Lancet 2024; 403:588-589. [PMID: 38342125 DOI: 10.1016/s0140-6736(23)02578-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 11/16/2023] [Indexed: 02/13/2024]
Affiliation(s)
- Ana Sofia Cerdeira
- Fetal Maternal Medicine Unit, Queen Charlotte's and Chelsea Hospital, London, UK; Nuffield Department of Women's Health and Reproductive Research, University of Oxford, Oxford OX3 9DU, UK.
| | | |
Collapse
|
146
|
Thadhani R, Cerdeira AS, Karumanchi SA. Translation of mechanistic advances in preeclampsia to the clinic: Long and winding road. FASEB J 2024; 38:e23441. [PMID: 38300220 DOI: 10.1096/fj.202301808r] [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: 09/05/2023] [Revised: 12/30/2023] [Accepted: 01/09/2024] [Indexed: 02/02/2024]
Abstract
As one of the leading causes of premature birth and maternal and infant mortality worldwide, preeclampsia remains a major unmet public health challenge. Preeclampsia and related hypertensive disorders of pregnancy are estimated to cause >75 000 maternal and 500 000 infant deaths globally each year. Because of rising rates of risk factors such as obesity, in vitro fertilization and advanced maternal age, the incidence of preeclampsia is going up with rates ranging from 5% to 10% of all pregnancies worldwide. A major discovery in the field was the realization that the clinical phenotypes related to preeclampsia, such as hypertension, proteinuria, and other adverse maternal/fetal events, are due to excess circulating soluble fms-like tyrosine kinase-1 (sFlt-1, also referred to as sVEGFR-1). sFlt-1 is an endogenous anti-angiogenic protein that is made by the placenta and acts by neutralizing the pro-angiogenic proteins vascular endothelial growth factor (VEGF) and placental growth factor (PlGF). During the last decade, this work has spawned a new era of molecular diagnostics for early detection of this condition. Antagonizing sFlt-1 either by reducing production or blocking its actions has shown salutary effects in animal models. Further, in early-stage human studies, the therapeutic removal of sFlt-1 from maternal circulation has shown promise in delaying disease progression and improving outcomes. Recently, the FDA approved the first molecular test for preterm preeclampsia (sFlt-1/PlGF ratio) for clinical use in the United States. Measuring serum sFlt-1/PlGF ratio in the acute hospital setting may aid short-term management, particularly regarding step-up or step-down of care, decision to transfer to settings better equipped to manage both the mother and the preterm neonate, appropriate timing of administration of steroids and magnesium sulfate, and in expectant management decisions. The test itself has the potential to save lives. Furthermore, the availability of a molecular test that correlates with adverse outcomes has set the stage for interventional clinical trials testing treatments for this disorder. In this review, we will discuss the role of circulating sFlt-1 and related factors in the pathogenesis of preeclampsia and specifically how this discovery is leading to concrete advances in the care of women with preeclampsia.
Collapse
Affiliation(s)
- Ravi Thadhani
- Woodruff Health Sciences Center, Emory University School of Medicine, Atlanta, Georgia, USA
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Ana Sofia Cerdeira
- Nuffield Department of Women's Health and Reproductive Research, University of Oxford, Oxford, UK
- Fetal Maternal Medicine Unit, Queen Charlotte's and Chelsea Hospital, London, UK
| | - S Ananth Karumanchi
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
147
|
Dijmărescu AL, Tănase F, Novac MB, Siminel MA, Rotaru I, Caragea DC, Manolea MM, Văduva CC, Boldeanu MV, Boldeanu L. Longitudinal 8-Epi-Prostaglandin F2-Alpha and Angiogenic Profile Mediator Evaluation during Pregnancy in Women with Suspected or Confirmed Pre-eclampsia. Biomedicines 2024; 12:433. [PMID: 38398035 PMCID: PMC10886743 DOI: 10.3390/biomedicines12020433] [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: 12/19/2023] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Background: In this exploratory study, we aimed to evaluate the dynamics of angiogenic [soluble fms-like tyrosine kinase-1 (sFlt-1), placental growth factor (PlGF), soluble Endoglin (sEng), and sFlt-1/PlGF, PlGF/sFlt-1, and sEng/PlGF ratios] and oxidative stress [8-epi-prostaglandin F2 alpha (8-epi-PGF2α) and 8-epi-PGF2α/PlGF ratio] mediator levels in women with suspected or confirmed pre-eclampsia (PE) at least two times during pregnancy. We also wanted to identify the possible correlations between 8-epi-PGF2α and angiogenic mediator levels at the time of inclusion of pregnant women. Methods: We included 40 pregnant women with suspected or confirmed PE, with a mean age of 29 years (range between 18 and 41 years) and gestational age between 18 and 28 weeks at inclusion in this study. The Enzyme-Linked Immunosorbent Assay (ELISA) method to measure the levels of serum angiogenic and oxidative stress mediators was used. Results: The evaluation of baseline sFlt-1/PlGF ratios using a cut-off of 38 suggested that 25 pregnant women had a sFlt-1/PlGF ratio of >38 (sFlt-1/PlGF ratio of >38 group) and 15 had a sFlt-1/PlGF ratio of ≤38 (sFlt-1/PlGF ratio of ≤38 group). The increases in sFlt-1/PlGF ratio in the sFlt-1/PlGF ratio of >38 group were caused by both an increase in sFlt-1 (2.04-fold) and a decrease in PlGF levels (2.55-fold). The 8-epi-PGF2α median levels were higher in the sFlt-1/PlGF ratio of >38 group (1.62-fold). During follow-up after pregnancy, we observed that the mean values of sFlt-1 and sEng and the median values of 8-epi-PGF2α and sFlt-1/PlGF, sEng/PlGF, and 8-epi-PGF2α/PlGF ratios increased directly proportional to gestational age for each measurement time until delivery in both groups. For five women who had a sFlt-1/PlGF ratio ≤38 at inclusion, sFlt-1/PlGF ratio was observed to increase to >38 later in pregnancy. We observed that, in the sFlt-1/PlGF ratio >38 group, baseline 8-epi-PGF2α levels better correlated with angiogenic mediator levels. Conclusions: Our study shows that 33.33% of pregnant women evaluated for suspected or confirmed PE with a sFlt-1/PlGF ratio of ≤38 displayed a rise in sFlt-1/PlGF ratio in subsequent weeks. In addition, together with angiogenic mediators, 8-epi-PGF2 α can be utilized as an independent predictor factor to help clinicians identify or predict which pregnant women will develop PE.
Collapse
Affiliation(s)
- Anda Lorena Dijmărescu
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (A.L.D.); (F.T.); (M.M.M.); (C.-C.V.)
| | - Florentina Tănase
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (A.L.D.); (F.T.); (M.M.M.); (C.-C.V.)
| | - Marius Bogdan Novac
- Department of Anesthesiology and Intensive Care, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Mirela Anişoara Siminel
- Department of Neonatology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Ionela Rotaru
- Department of Hematology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Daniel Cosmin Caragea
- Department of Nephrology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Maria Magdalena Manolea
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (A.L.D.); (F.T.); (M.M.M.); (C.-C.V.)
| | - Constantin-Cristian Văduva
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (A.L.D.); (F.T.); (M.M.M.); (C.-C.V.)
| | - Mihail Virgil Boldeanu
- Department of Immunology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Medico Science SRL—Stem Cell Bank Unit, 200690 Craiova, Romania
| | - Lidia Boldeanu
- Department of Microbiology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| |
Collapse
|
148
|
Giannubilo SR, Marzioni D, Tossetta G, Ciavattini A. HELLP Syndrome and Differential Diagnosis with Other Thrombotic Microangiopathies in Pregnancy. Diagnostics (Basel) 2024; 14:352. [PMID: 38396391 PMCID: PMC10887663 DOI: 10.3390/diagnostics14040352] [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: 12/31/2023] [Revised: 01/24/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
Thrombotic microangiopathies (TMAs) comprise a distinct group of diseases with different manifestations that can occur in both pediatric and adult patients. They can be hereditary or acquired, with subtle onset or a rapidly progressive course, and they are particularly known for their morbidity and mortality. Pregnancy is a high-risk time for the development of several types of thrombotic microangiopathies. The three major syndromes are hemolysis, elevated liver function tests, and low platelets (HELLP); hemolytic uremic syndrome (HUS); and thrombotic thrombocytopenic purpura (TTP). Because of their rarity, clinical information and therapeutic results related to these conditions are often obtained from case reports, small series, registries, and reviews. The collection of individual observations, the evolution of diagnostic laboratories that have identified autoimmune and/or genetic abnormalities using von Willebrand factor post-secretion processing or genetic-functional alterations in the regulation of alternative complement pathways in some of these TMAs, and, most importantly, the introduction of advanced treatments, have enabled the preservation of affected organs and improved survival rates. Although TMAs may show different etiopathogenesis routes, they all show the presence of pathological lesions, which are characterized by endothelial damage and the formation of thrombi rich in platelets at the microvascular level, as a common denominator, and thrombotic damage to microcirculation pathways induces "mechanical" (microangiopathic) hemolytic anemia, the consumption of platelets, and ischemic organ damage. In this review, we highlight the current knowledge about the diagnosis and management of these complications during pregnancy.
Collapse
Affiliation(s)
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Polytechnic University of Marche, Via Tronto 10/a, 60126 Ancona, Italy; (D.M.); (G.T.)
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Polytechnic University of Marche, Via Tronto 10/a, 60126 Ancona, Italy; (D.M.); (G.T.)
| | - Andrea Ciavattini
- Department of Clinical Sciences, Polytechnic University of Marche, Via Corridoni 11, 60123 Ancona, Italy;
| |
Collapse
|
149
|
Tóth E, Györffy D, Posta M, Hupuczi P, Balogh A, Szalai G, Orosz G, Orosz L, Szilágyi A, Oravecz O, Veress L, Nagy S, Török O, Murthi P, Erez O, Papp Z, Ács N, Than NG. Decreased Expression of Placental Proteins in Recurrent Pregnancy Loss: Functional Relevance and Diagnostic Value. Int J Mol Sci 2024; 25:1865. [PMID: 38339143 PMCID: PMC10855863 DOI: 10.3390/ijms25031865] [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] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Miscarriages affect 50-70% of all conceptions and 15-20% of clinically recognized pregnancies. Recurrent pregnancy loss (RPL, ≥2 miscarriages) affects 1-5% of recognized pregnancies. Nevertheless, our knowledge about the etiologies and pathophysiology of RPL is incomplete, and thus, reliable diagnostic/preventive tools are not yet available. Here, we aimed to define the diagnostic value of three placental proteins for RPL: human chorionic gonadotropin free beta-subunit (free-β-hCG), pregnancy-associated plasma protein-A (PAPP-A), and placental growth factor (PlGF). Blood samples were collected from women with RPL (n = 14) and controls undergoing elective termination of pregnancy (n = 30) at the time of surgery. Maternal serum protein concentrations were measured by BRAHMS KRYPTOR Analyzer. Daily multiple of median (dMoM) values were calculated for gestational age-specific normalization. To obtain classifiers, logistic regression analysis was performed, and ROC curves were calculated. There were differences in changes of maternal serum protein concentrations with advancing healthy gestation. Between 6 and 13 weeks, women with RPL had lower concentrations and dMoMs of free β-hCG, PAPP-A, and PlGF than controls. PAPP-A dMoM had the best discriminative properties (AUC = 0.880). Between 9 and 13 weeks, discriminative properties of all protein dMoMs were excellent (free β-hCG: AUC = 0.975; PAPP-A: AUC = 0.998; PlGF: AUC = 0.924). In conclusion, free-β-hCG and PAPP-A are valuable biomarkers for RPL, especially between 9 and 13 weeks. Their decreased concentrations indicate the deterioration of placental functions, while lower PlGF levels indicate problems with placental angiogenesis after 9 weeks.
Collapse
Affiliation(s)
- Eszter Tóth
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary
| | - Dániel Györffy
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary
| | - Máté Posta
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary
- Doctoral School, Semmelweis University, H-1085 Budapest, Hungary
| | - Petronella Hupuczi
- Maternity Private Clinic of Obstetrics and Gynecology, H-1126 Budapest, Hungary
| | - Andrea Balogh
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary
| | - Gábor Szalai
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary
- Department of Surgery, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Gergő Orosz
- Department of Obstetrics and Gynecology, Medical School, University of Debrecen, H-4032 Debrecen, Hungary
| | - László Orosz
- Department of Obstetrics and Gynecology, Medical School, University of Debrecen, H-4032 Debrecen, Hungary
| | - András Szilágyi
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary
| | - Orsolya Oravecz
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary
- Doctoral School of Biology, ELTE Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Lajos Veress
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Debrecen, H-4032 Debrecen, Hungary
| | - Sándor Nagy
- Faculty of Health and Sport Sciences, Széchenyi István University, H-9026 Győr, Hungary
| | - Olga Török
- Department of Obstetrics and Gynecology, Medical School, University of Debrecen, H-4032 Debrecen, Hungary
| | - Padma Murthi
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Clayton 3168, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Royal Women’s Hospital, Parkville 3052, Australia
| | - Offer Erez
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben Gurion University of the Negev, Be’er Sheva 8410501, Israel
- Department of Obstetrics and Gynecology, Medical School, Wayne State University, Detroit, MI 48201, USA
| | - Zoltán Papp
- Maternity Private Clinic of Obstetrics and Gynecology, H-1126 Budapest, Hungary
- Department of Obstetrics and Gynecology, Medical School, Semmelweis University, 27 Baross Street, H-1088 Budapest, Hungary
| | - Nándor Ács
- Department of Obstetrics and Gynecology, Medical School, Semmelweis University, 27 Baross Street, H-1088 Budapest, Hungary
| | - Nándor Gábor Than
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary
- Maternity Private Clinic of Obstetrics and Gynecology, H-1126 Budapest, Hungary
- Department of Obstetrics and Gynecology, Medical School, Semmelweis University, 27 Baross Street, H-1088 Budapest, Hungary
| |
Collapse
|
150
|
Lekva T, Michelsen AE, Roland MCP, Norwitz ER, Estensen ME, Olstad OK, Akkouh IA, Henriksen T, Bollerslev J, Aukrust P, Ueland T. Increased ferroptosis in leukocytes from preeclamptic women involving the long non-coding taurine upregulated gene 1 (TUG1). J Intern Med 2024; 295:181-195. [PMID: 37870937 DOI: 10.1111/joim.13732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
BACKGROUND Ferroptosis plays a key role in placental development and physiology, and abnormal ferroptosis has been implicated in trophoblast injury leading to preeclampsia (PE). We hypothesize that leukocytes isolated from PE exhibit increased ferroptosis and that extracellular vesicles contain long non-coding (lnc) RNA/mRNAs that modulate oxidative stress and iron toxicity in vascular endothelial cells. METHODS We measured the expression of key regulators of ferroptosis in leukocytes and extracellular vesicles as well as circulating biomarkers of iron homeostasis and oxidative stress in plasma from women with/without PE at different timepoints during pregnancy. For markers that were dysregulated, we assessed their temporal correlation with established markers of disease activity and marker of endothelial activation. For markers dysregulated in early pregnancy, we assessed their ability to predict the development of PE. RESULTS We found decreased lncRNA/mRNAs in leukocytes, but not extracellular vesicles, in PE that may modulate oxidative stress and iron toxicity. This decrease in anti-ferroptotic markers does not appear to be related to maternal disease activity or plasma oxidative stress status but rather to attenuated anti-inflammatory expression in these cells. Circulating ferritin was elevated in PE, supporting the hypothesis that PE represents a disbalance in iron homeostasis. Low lncRNA taurine upregulated gene 1 RNA levels in leukocytes at 22-24 weeks were strongly associated with the development of PE. CONCLUSIONS Our findings suggest that maternal leukocytes in PE show decreased anti-ferroptotic activity that correlates with anti-inflammatory expression. Moreover, some of these changes in ferroptotic activity appear to precede the development of PE.
Collapse
Affiliation(s)
- Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Annika Elisabet Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - Errol R Norwitz
- Newton-Wellesley Hospital and Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | - Ole Kristoffer Olstad
- The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Ibrahim A Akkouh
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Tore Henriksen
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Obstetrics, Oslo University Hospital, Oslo, Norway
| | - Jens Bollerslev
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|