1
|
Tian Y, Liu M, Sun JY, Wang Y, Chen L, Sun W, Zhou L. Diagnosis of preeclampsia using metabolomic biomarkers. Hypertens Pregnancy 2024; 43:2379386. [PMID: 39039822 DOI: 10.1080/10641955.2024.2379386] [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: 04/19/2024] [Accepted: 06/30/2024] [Indexed: 07/24/2024]
Abstract
The diagnostic criteria for preeclampsia do not accurately reflect the pathophysiological characteristics of patients with preeclampsia. Conventional biomarkers and diagnostic approaches have proven insufficient to fully comprehend the intricacies of preeclampsia. This study aimed to screen differentially abundant metabolites as candidate biomarkers for preeclampsia. A propensity score matching method was used to perform a 1:1 match between preeclampsia patients (n = 70) and healthy control individuals (n = 70). Based on univariate and multivariate statistical analysis methods, the different characteristic metabolites were screened and identified. Least absolute shrinkage and selection operator (LASSO) regression analysis was subsequently used to further screen for differentially abundant metabolites. A receiver operating characteristic (ROC) curve was drawn to evaluate the diagnostic efficacy of the metabolites. A total of 1,630 metabolites were identified and quantified in maternal serum samples. Fifty-three metabolites were significantly increased, and two were significantly decreased in preeclampsia patients. The area under the curve (AUC) of the model composed of isobutyryl-L-carnitine and acetyl-leucine was 0.878, and the sensitivity and specificity in detecting preeclampsia were 81.4% and 87.1%, respectively. There are significant differences in metabolism between preeclampsia patients and healthy pregnant women, and a range of novel biomarkers have been identified. These findings lay the foundation for the use of metabolomic biomarkers for the diagnosis of preeclampsia.
Collapse
Affiliation(s)
- Yunfan Tian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mingwei Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jin-Yu Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yifeng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lianmin Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ling Zhou
- Department of Obstetrics and Gynecology, Liyang People's Hospital, Liyang, Jiangsu, China
| |
Collapse
|
2
|
Siboto A, Ludidi A, Sibiya N, Khathi A, Ngubane P. Maternal prediabetes as a risk factor of preeclampsia and placental dysfunction in pregnant female Sprague-Dawley rats. J OBSTET GYNAECOL 2024; 44:2379498. [PMID: 39084241 DOI: 10.1080/01443615.2024.2379498] [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: 07/28/2023] [Accepted: 06/24/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Prediabetes (PD) is associated with intermediate hyperglycaemia, dyslipidaemia, reduced nitric oxide (NO) bioavailability and moderate hypertension. All these factors are risk factor for preeclampsia (PE). However, the effects of the PD on placental function have not been shown. Accordingly, this study sought to investigate a possible link between maternal PD and the risk of developing PE. METHODS Pregnant female Sprague-Dawley rats (N = 18) were divided into normal, preeclamptic and prediabetic groups (n = 6 in each group) to study the effects of maternal PD on placenta function over the period of 19 days. Blood glucose and blood pressure were measured on gestational day (GND) 0, 9 and 18. Placental vascular endothelial growth factor (VEGF), placenta growth factor (PlGF) and soluble fms-like tyrosine kinase 1 (sFlt-1) mRNA expression were measured terminally. Data were analysed using ANOVA followed by the Tukey-Kramer post hoc test. Values of p < .05 were used to indicate statistical significance. RESULTS Maternal PD and PE significantly increased blood glucose, decrease NO concentration and increase in MAP by comparison to the normal pregnant control group. Maternal PD significantly decreased VEGF, PlGF mRNA expression with a slight increase in sFlt-1 mRNA expression comparison to the normal pregnant control group. CONCLUSIONS Maternal PD is associated with placental dysfunction due to impaired glucose handling, endothelial dysfunction and an imbalance in angiogenic and antiangiogenic factors. Therefore, maternal PD is a risk factor of PE.
Collapse
Affiliation(s)
- Aneliswe Siboto
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu Natal, Durban, South Africa
| | - Asiphaphola Ludidi
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu Natal, Durban, South Africa
| | - Ntethelelo Sibiya
- Division of Pharmacology, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
| | - Andile Khathi
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu Natal, Durban, South Africa
| | - Phikelelani Ngubane
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu Natal, Durban, South Africa
| |
Collapse
|
3
|
Li C, Sheng J, Zhang Y, Lyu Q, Yang L, Zhong Z. Thyroid function and preeclampsia: a two-sample bidirectional Mendelian randomization study. J Hypertens 2024; 42:2075-2083. [PMID: 39248096 DOI: 10.1097/hjh.0000000000003791] [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: 03/17/2024] [Accepted: 06/03/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Thyroid dysfunction has been associated with preeclampsia (PE) during pregnancy, but the observational results are conflicting. Our study aims to investigate the causal association and direction between genetically predicted effects of thyroid function on PE and vice versa via two large summary genetic data. METHODS We conducted a two-sample bidirectional Mendelian randomization (MR) study using genome-wide association studies (GWAS) summary data from two primarily European cohorts: the ThyroidOmics Consortium and the FinnGen Biobank. We applied the random effects inverse variance weighted (IVW) as our main analysis. MR-Egger and weighted median were used for sensitivity analysis. Statistical analysis was performed using the R program (version 4.3.0) with the two-sample package (version 0.5.6). RESULTS The results suggest that genetically predicted hyperthyroidism is causally associated with PE during pregnancy [ β = 0.06, 95% confidence interval (CI): 1.01-1.12; P = 0.02], and genetically predicted hypothyroidism is also causally associated with PE during pregnancy ( β = 0.11, 95% CI: 1.03-1.21; P = 0.01). These effects were further confirmed with sensitivity analysis. Conversely, preeclampsia is not associated with the risk of thyroid dysfunction in the reverse MR results: thyroid-stimulating hormone ( β = 0.00, P = 0.92), free thyroxine (FT4) ( β = -0.01, P = 0.56), triiodothyronine (FT3) ( β = -0.00, P = 0.72), FT3/FT4 ( β = -0.01, P = 0.38), thyroid peroxidase antibodies ( β = -0.01, P = 0.64), hyperthyroidism ( β = -0.11, P = 0.29) and hypothyroidism ( β = 0.04, P = 0.12). CONCLUSION Our study suggests that hyper-/hypo-thyroidism causally affected preeclampsia, while PE is not causally associated with thyroid dysfunctions.
Collapse
Affiliation(s)
- Chu Li
- Center for Reproductive Medicine, Department of Obstetrics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College)
- The Second Clinical College of Zhejiang Chinese Medical University, Hangzhou
| | - Jingjing Sheng
- Department of Obstetrics and Gynecology, the Fourth Affiliated Hospital, College of Medicine, Zhejiang University, Yiwu, China
| | - Yawei Zhang
- Center for Reproductive Medicine, Department of Obstetrics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College)
- The Second Clinical College of Zhejiang Chinese Medical University, Hangzhou
| | - Qiaofei Lyu
- Center for Reproductive Medicine, Department of Obstetrics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College)
- The Second Clinical College of Zhejiang Chinese Medical University, Hangzhou
| | - Liwei Yang
- Center for Reproductive Medicine, Department of Obstetrics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College)
| | - Zixing Zhong
- Center for Reproductive Medicine, Department of Obstetrics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College)
| |
Collapse
|
4
|
Schor JS, Kadambi A, Fulcher I, Venkatesh KK, Clapp MA, Ebrahim S, Ebrahim A, Wen T. Using machine learning to predict the risk of developing hypertensive disorders of pregnancy using a contemporary nulliparous cohort. AJOG GLOBAL REPORTS 2024; 4:100386. [PMID: 39385801 PMCID: PMC11462053 DOI: 10.1016/j.xagr.2024.100386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024] Open
Abstract
Background Hypertensive disorders of pregnancy (HDP) are significant drivers of maternal and neonatal morbidity and mortality. Current management strategies include early identification and initiation of risk mitigating interventions facilitated by a rules-based checklist. Advanced analytic techniques, such as machine learning, can potentially offer improved and refined predictive capabilities. Objective To develop and internally validate a machine learning prediction model for hypertensive disorders of pregnancy (HDP) when initiating prenatal care. Study Design We developed a prediction model using data from the prospective multisite cohort Nulliparous Pregnancy Outcomes Study: Monitoring Mothers-to-Be (nuMoM2b) among low-risk individuals without a prior history of aspirin utilization for preeclampsia prevention. The primary outcome was the development of HDP. Random forest modeling was utilized to develop predictive models. Recursive feature elimination (RFE) was employed to create a reduced model for each outcome. Area under the curve (AUC), 95% confidence intervals (CI), and calibration curves were utilized to assess discrimination and accuracy. Sensitivity analyses were conducted to compare the sensitivity and specificity of the reduced model compared to existing risk factor-based algorithms. Results Of 9,124 assessed low risk nulliparous individuals, 21% (n=1,927) developed HDP. The prediction model for HDP had satisfactory discrimination with an AUC of 0.73 (95% CI: 0.70, 0.75). After RFE, a parsimonious reduced model with 30 features was created with an AUC of 0.71 (95% CI: 0.68, 0.74). Variables included in the model after RFE included body mass index at the first study visit, pre-pregnancy weight, first trimester complete blood count results, and maximum systolic blood pressure at the first visit. Calibration curves for all models revealed relatively stable agreement between predicted and observed probabilities. Sensitivity analysis noted superior sensitivity (AUC 0.80 vs 0.65) and specificity (0.65 vs 0.53) of the model compared to traditional risk factor-based algorithms. Conclusion In cohort of low-risk nulliparous pregnant individuals, a prediction model may accurately predict HDP diagnosis at the time of initiating prenatal care and aid employment of close interval monitoring and prophylactic measures earlier in pregnancy.
Collapse
Affiliation(s)
- Jonathan S. Schor
- Delfina Care Inc, San Francisco, CA, USA (Schor, Kadambi, Fulcher, Venkatesh, Clapp, Ebrahim, Ebrahim and Wen)
- University of California, San Francisco (UCSF) Medical Scientist Training Program, San Francisco, CA, USA (Schor)
| | - Adesh Kadambi
- Delfina Care Inc, San Francisco, CA, USA (Schor, Kadambi, Fulcher, Venkatesh, Clapp, Ebrahim, Ebrahim and Wen)
- Department of Biomedical Engineering, University of Toronto, Toronto, Canada (Kadambi)
| | - Isabel Fulcher
- Delfina Care Inc, San Francisco, CA, USA (Schor, Kadambi, Fulcher, Venkatesh, Clapp, Ebrahim, Ebrahim and Wen)
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, Massachusetts, USA (Fulcher)
| | - Kartik K. Venkatesh
- Delfina Care Inc, San Francisco, CA, USA (Schor, Kadambi, Fulcher, Venkatesh, Clapp, Ebrahim, Ebrahim and Wen)
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Ohio State University Medical Center, Columbus, OH, USA (Venkatesh)
| | - Mark A. Clapp
- Delfina Care Inc, San Francisco, CA, USA (Schor, Kadambi, Fulcher, Venkatesh, Clapp, Ebrahim, Ebrahim and Wen)
| | - Senan Ebrahim
- Delfina Care Inc, San Francisco, CA, USA (Schor, Kadambi, Fulcher, Venkatesh, Clapp, Ebrahim, Ebrahim and Wen)
| | - Ali Ebrahim
- Delfina Care Inc, San Francisco, CA, USA (Schor, Kadambi, Fulcher, Venkatesh, Clapp, Ebrahim, Ebrahim and Wen)
| | - Timothy Wen
- Delfina Care Inc, San Francisco, CA, USA (Schor, Kadambi, Fulcher, Venkatesh, Clapp, Ebrahim, Ebrahim and Wen)
- Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco (UCSF), San Francisco, CA USA (Wen)
| |
Collapse
|
5
|
Melamed N, Kingdom JC, Fu L, Yip PM, Arruda-Caycho I, Hui D, Hladunewich MA. Predictive and Diagnostic Value of the Angiogenic Proteins in Patients With Chronic Kidney Disease. Hypertension 2024; 81:2251-2262. [PMID: 39162032 DOI: 10.1161/hypertensionaha.124.23411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/30/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Our objective was to investigate the predictive and diagnostic accuracy of the angiogenic proteins sFlt-1 (soluble fms-like tyrosine kinase-1) and PlGF (placental growth factor) for preterm preeclampsia and explore the relationship between renal function and these proteins. METHODS We completed a blinded, prospective, longitudinal, observational study of patients with chronic kidney disease followed at a tertiary center (2018-2023). Serum samples were obtained at 3 time points along gestation (planned sampling): 12-16, 18-22, and 28-32 weeks. In addition, samples were obtained whenever preeclampsia was suspected (indicated sampling). sFlt-1 and PlGF levels remained concealed until the study ended. The primary outcome was preterm preeclampsia. The planned and indicated samples were used to estimate the predictive and diagnostic accuracy of the angiogenic proteins, respectively. RESULTS Of the 97 participants, 21 (21.6%) experienced preterm preeclampsia. In asymptomatic patients with chronic kidney disease, the angiogenic proteins were predictive of preterm preeclampsia only when sampled in the third trimester, in which case the sFlt-1/PlGF ratio (false positive rate of 37% for a detection rate of 80%) was more predictive than either sFlt-1 or PlGF in isolation. In patients with suspected preeclampsia, the diagnostic accuracy of the sFlt-1/PlGF ratio (false positive rate of 26% for a detection rate of 80%) was higher than that of sFlt-1 and PlGF in isolation. Diminished renal function was associated with increased levels of PlGF. CONCLUSIONS sFlt-1 and PlGF can effectively predict and improve the diagnostic accuracy for preterm preeclampsia among patients with chronic kidney disease. The optimal sFlt-1/PlGF ratio cutoff to rule out preeclampsia may need to be lower in patients with impaired renal function.
Collapse
Affiliation(s)
- Nir Melamed
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, Temerty Faculty of Medicine (N.M., I.A.-C., D.H.), University of Toronto, Ontario, Canada
| | - John C Kingdom
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Mount Sinai Hospital, Temerty Faculty of Medicine (J.C.K.), University of Toronto, Ontario, Canada
| | - Lei Fu
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre (L.F., P.M.Y.), University of Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology (L.F., P.M.Y.), University of Toronto, Ontario, Canada
| | - Paul M Yip
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre (L.F., P.M.Y.), University of Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology (L.F., P.M.Y.), University of Toronto, Ontario, Canada
| | - Isabel Arruda-Caycho
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, Temerty Faculty of Medicine (N.M., I.A.-C., D.H.), University of Toronto, Ontario, Canada
| | - Dini Hui
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, Temerty Faculty of Medicine (N.M., I.A.-C., D.H.), University of Toronto, Ontario, Canada
| | - Michelle A Hladunewich
- Division of Nephrology and Obstetric Medicine, Department of Medicine, Sunnybrook Health Sciences Centre, Temerty Faculty of Medicine (M.A.H.), University of Toronto, Ontario, Canada
| |
Collapse
|
6
|
Ricart M, Bonacina E, Garcia‐Manau P, López M, Caamiña S, Vives À, Lopez‐Quesada E, Maroto A, de Mingo L, Pintado E, Ferrer‐Costa R, Martín L, Rodriguez‐Zurita A, Garcia E, Pallarols M, Pratcorona L, Teixidor M, Orizales‐Lago C, Ocaña V, del Barco E, Carreras E, Suy A, Mendoza M. Placental growth factor at 24-28 weeks for aspirin discontinuation in pregnancies at high risk for preterm preeclampsia: Post hoc analysis of StopPRE trial. Acta Obstet Gynecol Scand 2024; 103:2273-2280. [PMID: 39171611 PMCID: PMC11502455 DOI: 10.1111/aogs.14955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024]
Abstract
INTRODUCTION This study aims to evaluate the safety of discontinuing aspirin treatment at 24-28 weeks in women at high risk after first-trimester combined screening for preeclampsia (PE) and normal placental growth factor (PlGF) levels at 24-28 weeks of gestation. MATERIAL AND METHODS This is a post hoc analysis of the StopPRE trial, conducted at nine Spanish maternity hospitals from September 2019 to September 2021. In the StopPRE trial, all high-risk single pregnancies identified during first-trimester screening for PE were treated with 150 mg of daily aspirin. Out of 1604 eligible women with a soluble fms-like tyrosine kinase-1 to PlGF ratio (sFlt-1/PlGF) ≤38 at 24-28 weeks, 968 were randomly assigned in a 1:1 ratio to either continue aspirin until 36 weeks (control group) or discontinue it (intervention group). In this secondary analysis, only women with PlGF ≥100 pg/mL at 24-28 weeks were included. As in the StopPRE trial, the non-inferiority margin was set at a 1.9% difference in preterm PE incidence between the groups. RESULTS Among the 13 983 screened pregnant women, 1984 (14.2%) were deemed high-risk for preterm PE, of which 397 (20.0%) were ineligible, 636 declined participation, and 32 were excluded. Ultimately, 919 women with PlGF >100 pg/mL were randomized and included in this analysis. Preterm PE occurred in 0.9% of the intervention group (4 out of 465) and 1.5% of the control group (7 out of 454), indicating non-inferiority of aspirin discontinuation. There were no significant differences between the groups in adverse pregnancy outcomes before 37 weeks, at <34 weeks, or ≥37 weeks. Minor antepartum hemorrhage incidence was significantly lower in the intervention group (absolute difference, -5.96; 95% CI, -10.10 to -1.82). CONCLUSIONS Discontinuation of aspirin treatment at 24-28 weeks in women with PlGF levels ≥100 pg/mL was non-inferior to continuing until 36 weeks for preventing preterm PE. However, these findings should be interpreted with caution, as they originate from a subanalysis of the StopPRE trial.
Collapse
Affiliation(s)
- Marta Ricart
- Department of Pediatrics, Obstetrics and Gynecology, and Preventive Medicine and Public HealthUniversitat Autònoma de BarcelonaBellaterraSpain
- Department of ObstetricsHospital Universitari Germans Trias i PujolBadalonaSpain
| | - Erika Bonacina
- Maternal Fetal Medicine Unit, Department of ObstetricsVall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| | - Pablo Garcia‐Manau
- Maternal Fetal Medicine Unit, Department of ObstetricsVall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| | - Monica López
- Department of ObstetricsHospital Universitari de Tarragona Joan XXIIITarragonaSpain
| | - Sara Caamiña
- Department of ObstetricsHospital Universitario Nuestra Señora de CandelariaSanta Cruz de TenerifeSpain
| | - Àngels Vives
- Department of ObstetricsConsorci Sanitari de TerrassaTerrassaSpain
| | - Eva Lopez‐Quesada
- Department of ObstetricsHospital Universitari Mútua TerrassaTerrassaSpain
| | - Anna Maroto
- Department of ObstetricsHospital Universitari de Girona Doctor Josep TruetaGironaSpain
| | - Laura de Mingo
- Department of ObstetricsHospital Universitario Severo OchoaLeganésSpain
| | - Elena Pintado
- Department of ObstetricsHospital Universitario de GetafeGetafeSpain
| | - Roser Ferrer‐Costa
- Department of BiochemistryVall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| | - Lourdes Martín
- Department of ObstetricsHospital Universitari de Tarragona Joan XXIIITarragonaSpain
| | - Alicia Rodriguez‐Zurita
- Department of ObstetricsHospital Universitario Nuestra Señora de CandelariaSanta Cruz de TenerifeSpain
| | - Esperanza Garcia
- Department of ObstetricsConsorci Sanitari de TerrassaTerrassaSpain
| | - Mar Pallarols
- Department of ObstetricsHospital Universitari Mútua TerrassaTerrassaSpain
| | - Laia Pratcorona
- Department of Pediatrics, Obstetrics and Gynecology, and Preventive Medicine and Public HealthUniversitat Autònoma de BarcelonaBellaterraSpain
- Department of ObstetricsHospital Universitari Germans Trias i PujolBadalonaSpain
| | - Mireia Teixidor
- Department of ObstetricsHospital Universitari de Girona Doctor Josep TruetaGironaSpain
| | | | - Vanesa Ocaña
- Department of ObstetricsHospital Universitario de GetafeGetafeSpain
| | - Esther del Barco
- Maternal Fetal Medicine Unit, Department of ObstetricsVall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| | - Elena Carreras
- Maternal Fetal Medicine Unit, Department of ObstetricsVall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| | - Anna Suy
- Department of Pediatrics, Obstetrics and Gynecology, and Preventive Medicine and Public HealthUniversitat Autònoma de BarcelonaBellaterraSpain
- Maternal Fetal Medicine Unit, Department of ObstetricsVall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| | - Manel Mendoza
- Department of Pediatrics, Obstetrics and Gynecology, and Preventive Medicine and Public HealthUniversitat Autònoma de BarcelonaBellaterraSpain
- Maternal Fetal Medicine Unit, Department of ObstetricsVall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| |
Collapse
|
7
|
Novoa RH, Pérez-Aliaga C, Castañeda-Apolinario JE, Ramírez-Moreno AI, Meza-Santibañez L. Screening, prevention and early diagnosis of preeclampsia: need for an updated protocol in Peru. Rev Peru Med Exp Salud Publica 2024; 41:321-323. [PMID: 39442116 PMCID: PMC11495920 DOI: 10.17843/rpmesp.2024.413.13793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/05/2024] [Indexed: 10/25/2024] Open
Affiliation(s)
- Rommy H. Novoa
- National Maternal Perinatal Institute. Lima, Peru.National Maternal Perinatal InstituteLimaPeru
| | - Carlos Pérez-Aliaga
- National Maternal Perinatal Institute. Lima, Peru.National Maternal Perinatal InstituteLimaPeru
| | - Jose E. Castañeda-Apolinario
- Alberto Hurtado Faculty of Medicine, Universidad Peruana Cayetano Heredia. Lima, Peru.Universidad Peruana Cayetano HerediaAlberto Hurtado Faculty of MedicineUniversidad Peruana Cayetano HerediaLimaPeru
| | - Alexandra I. Ramírez-Moreno
- Alberto Hurtado Faculty of Medicine, Universidad Peruana Cayetano Heredia. Lima, Peru.Universidad Peruana Cayetano HerediaAlberto Hurtado Faculty of MedicineUniversidad Peruana Cayetano HerediaLimaPeru
| | - Luis Meza-Santibañez
- National Maternal Perinatal Institute. Lima, Peru.National Maternal Perinatal InstituteLimaPeru
- San Fernando Faculty of Medicine, Universidad Nacional Mayor de San Marcos. Lima, Peru.Universidad Nacional Mayor de San MarcosSan Fernando Faculty of MedicineUniversidad Nacional Mayor de San MarcosLimaPeru
| |
Collapse
|
8
|
Dricot CEMK, Erreygers I, Cauwenberghs E, De Paz J, Spacova I, Verhoeven V, Ahannach S, Lebeer S. Riboflavin for women's health and emerging microbiome strategies. NPJ Biofilms Microbiomes 2024; 10:107. [PMID: 39420006 PMCID: PMC11486906 DOI: 10.1038/s41522-024-00579-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024] Open
Abstract
Riboflavin (vitamin B2) is an essential water-soluble vitamin that serves as a precursor of flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD). FMN and FAD are coenzymes involved in key enzymatic reactions in energy metabolism, biosynthesis, detoxification and electron scavenging pathways. Riboflavin deficiency is prevalent worldwide and impacts women's health due to riboflavin demands linked to urogenital and reproductive health, hormonal fluctuations during the menstrual cycle, pregnancy, and breastfeeding. Innovative functional foods and nutraceuticals are increasingly developed to meet women's riboflavin needs to supplement dietary sources. An emerging and particularly promising strategy is the administration of riboflavin-producing lactic acid bacteria, combining the health benefits of riboflavin with those of probiotics and in situ riboflavin production. Specific taxa of lactobacilli are of particular interest for women, because of the crucial role of Lactobacillus species in the vagina and the documented health effects of other Lactobacillaceae taxa in the gut and on the skin. In this narrative review, we synthesize the underlying molecular mechanisms and clinical benefits of riboflavin intake for women's health, and evaluate the synergistic potential of riboflavin-producing lactobacilli and other microbiota.
Collapse
Affiliation(s)
- Caroline E M K Dricot
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Isabel Erreygers
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Eline Cauwenberghs
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Jocelyn De Paz
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Irina Spacova
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Veronique Verhoeven
- Department of Family Medicine and Population Health, University of Antwerp, Antwerp, Belgium
- U-MaMi Excellence Centre, University of Antwerp, Antwerp, Belgium
| | - Sarah Ahannach
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Sarah Lebeer
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium.
- U-MaMi Excellence Centre, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
9
|
Forrest M, Matossian M, Valdes Sustaita B, Papacostas Quintanilla H, Spronck B, Sharman J, Daskalopoulou SS. Arterial stiffness as a novel tool for the early prediction of preeclampsia: a perspective. J Hum Hypertens 2024:10.1038/s41371-024-00967-6. [PMID: 39415051 DOI: 10.1038/s41371-024-00967-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
Preeclampsia is a leading complication of pregnancy that lacks accurate tools for its early prediction. Improved risk stratification tools early in pregnancy would enable more efficient allocation of limited healthcare resources while ensuring that pregnant women destined to develop preeclampsia receive appropriate care. This brief perspective highlights the current state of first-trimester preeclampsia prediction. We focus on arterial stiffness, an important hemodynamic indicator of vascular health that has shown promising results for improved early prediction of preeclampsia by our and independent research groups. Further, we outline the promise, applicability, and feasibility of integrating arterial stiffness assessments into clinical practice.
Collapse
Affiliation(s)
- Mekayla Forrest
- Vascular Health Unit, Research Institute of McGill University Health Centre (RI-MUHC), Montreal, QC, Canada
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Maria Matossian
- Vascular Health Unit, Research Institute of McGill University Health Centre (RI-MUHC), Montreal, QC, Canada
| | - Brenda Valdes Sustaita
- Vascular Health Unit, Research Institute of McGill University Health Centre (RI-MUHC), Montreal, QC, Canada
- Department of Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Helena Papacostas Quintanilla
- Vascular Health Unit, Research Institute of McGill University Health Centre (RI-MUHC), Montreal, QC, Canada
- Department of Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Bart Spronck
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| | - James Sharman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Stella S Daskalopoulou
- Vascular Health Unit, Research Institute of McGill University Health Centre (RI-MUHC), Montreal, QC, Canada.
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada.
- Department of Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada.
- Division of Internal Medicine, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
10
|
Kim T, Choodinatha HK, Kim KS, Shin K, Kim HJ, Park JY, Hong JW, Lee LP. Understanding the role of soluble proteins and exosomes in non-invasive urine-based diagnosis of preeclampsia. Sci Rep 2024; 14:24117. [PMID: 39406891 PMCID: PMC11482518 DOI: 10.1038/s41598-024-75080-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
Preeclampsia is a hypertensive disorder of pregnancy that can lead to stillbirth and preterm birth if not treated promptly. Currently, the diagnosis of preeclampsia relies on clinical symptoms such as hypertension and proteinuria, along with invasive blood tests. Here, we investigate the role of soluble proteins and exosomes in noninvasive diagnosing preeclampsia non-invasively using maternal urine and urine-derived exosomes. We quantified the levels of particles and the presence of TSG101 and CD63 in urine and urinary exosomes via the biologically intact exosome separation technology (BEST) platform. Then, we obtained higher levels of soluble proteins such as fms-like tyrosine kinase-1 (sFlt-1) and placental growth factor (PlGF) from urine as it was than urinary exosomes. Compared to commercial blood tests, the sensitivity of the sFlt-1/PlGF ratio was found to be 4.0 times higher in urine tests and 1.5 times higher in tests utilizing urine-derived exosomes. Our findings offer promising possibilities for the early and non-invasive identification of high-risk individuals at risk of preeclampsia, allowing for comprehensive preventive management.
Collapse
Affiliation(s)
- Taewoon Kim
- Department of Bionanotechnology, Graduate School, Hanyang University, Seoul, 04763, Korea
| | - Harshitha Kallubhavi Choodinatha
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
- Seoul National University, Seoul, Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kwang Sik Kim
- Department of Bionanotechnology, Graduate School, Hanyang University, Seoul, 04763, Korea
| | - Kyusoon Shin
- Department of Bionanotechnology, Graduate School, Hanyang University, Seoul, 04763, Korea
| | - Hyeon Ji Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
- Seoul National University, Seoul, Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jee Yoon Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea.
- Seoul National University, Seoul, Korea.
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.
| | - Jong Wook Hong
- Department of Bionanotechnology, Graduate School, Hanyang University, Seoul, 04763, Korea.
- Department of Medical and Digital Engineering, Graduate School, Hanyang University, Seoul, 04763, Korea.
- Department of Bionanoengineering, Hanyang University, 15588, Ansan, Gyeonggi-do, Korea.
| | - Luke P Lee
- Harvard Medical School, Department of Medicine, Harvard University, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Bioengineering, University of California at Berkeley, Berkeley, CA, USA.
- Department of Electrical Engineering and Computer Science, University of California at Berkeley, Berkeley, CA, USA.
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Korea.
- Department of Chemistry & Nanoscience, Ewha Womans University, Seoul, Korea.
| |
Collapse
|
11
|
McEvoy JW, McCarthy CP, Bruno RM, Brouwers S, Canavan MD, Ceconi C, Christodorescu RM, Daskalopoulou SS, Ferro CJ, Gerdts E, Hanssen H, Harris J, Lauder L, McManus RJ, Molloy GJ, Rahimi K, Regitz-Zagrosek V, Rossi GP, Sandset EC, Scheenaerts B, Staessen JA, Uchmanowicz I, Volterrani M, Touyz RM. 2024 ESC Guidelines for the management of elevated blood pressure and hypertension. Eur Heart J 2024; 45:3912-4018. [PMID: 39210715 DOI: 10.1093/eurheartj/ehae178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
|
12
|
Jang W, Song EL, Mun SJ, Bong KW. Efficient isolation of encoded microparticles in a degassed micromold for highly sensitive and multiplex immunoassay with signal amplification. Biosens Bioelectron 2024; 261:116465. [PMID: 38850735 DOI: 10.1016/j.bios.2024.116465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/29/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
Multiplex detection of low-abundance protein biomarkers in biofluids can contribute to diverse biomedical fields such as early diagnosis and precision medicine. However, conventional techniques such as digital ELISA, microarray, and hydrogel-based assay still face limitations in terms of efficient protein detection due to issues with multiplexing capability, sensitivity, or complicated assay procedures. In this study, we present the degassed micromold-based particle isolation technique for highly sensitive and multiplex immunoassay with enzymatic signal amplification. Using degassing treatment of nanoporous polydimethylsiloxane (PDMS) micromold, the encoded particles are isolated in the mold within 5 min absorbing trapped air bubbles into the mold by air suction capability. Through 10 min of signal amplification in the isolated spaces by fluorogenic substrate and horseradish peroxidase labeled in the particle, the assay signal is amplified with one order of magnitude compared to that of the standard hydrogel-based assay. Using the signal amplification assay, vascular endothelial growth factor (VEGF) and chorionic gonadotropin beta (CG beta), the preeclampsia-related protein biomarkers, are quantitatively detected with a limit of detection (LoD) of 249 fg/mL and 476 fg/mL in phosphate buffer saline. The multiplex immunoassay is conducted to validate negligible non-specific detection signals and robust recovery rates in the multiplex assay. Finally, the VEGF and CG beta in real urine samples are simultaneously and quantitatively detected by the developed assay. Given the high sensitivity, multiplexing capability, and process simplicity, the presented particle isolation-based signal amplification assay holds significant potential in biomedical and proteomic fields.
Collapse
Affiliation(s)
- Wookyoung Jang
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - E Loomee Song
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Seok Joon Mun
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Ki Wan Bong
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
13
|
Jiang H, Meng T, Li Z. Role of circular RNAs in preeclampsia (Review). Exp Ther Med 2024; 28:372. [PMID: 39091629 PMCID: PMC11292168 DOI: 10.3892/etm.2024.12661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Preeclampsia (PE) is a hypertensive disorder of pregnancy characterized by new-onset hypertension and proteinuria after 20 weeks of gestation, which affects 3-8% of pregnant individuals worldwide each year. Prevention, diagnosis and treatment of PE are some of the most important problems faced by obstetrics. There is growing evidence that circular RNAs (circRNAs) are involved in the pathogenesis of PE. The present review summarizes the research progress of circRNAs and then describes the expression patterns of circRNAs in PE and their functional mechanisms affecting PE development. The role of circRNAs as biomarkers for the diagnosis of PE, and the research status of circRNAs in PE are summarized in the hope of finding novel strategies for the prevention and treatment of PE.
Collapse
Affiliation(s)
- Hengxue Jiang
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
- Department of Obstetrics and Gynecology, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Tao Meng
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ziwei Li
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
14
|
Wu H, Zhao L. Circ_0015382 Regulates the miR-942-5p/NDRG1 Axis to Suppress Trophoblast Cell Functions. Reprod Sci 2024:10.1007/s43032-024-01685-7. [PMID: 39354288 DOI: 10.1007/s43032-024-01685-7] [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: 05/13/2024] [Accepted: 08/18/2024] [Indexed: 10/03/2024]
Abstract
Circular RNAs (circRNAs) are non-coding RNAs that exert vital function in many human diseases, including preeclampsia (PE). Circ_0015382 is considered to be a key regulator of PE progression, so more molecular mechanisms need to be further studied. Real-time quantitative PCR was used to detect the mRNA levels of circ_0015382, miR-942-5p, and N-myc downstream regulated 1 (NDRG1). Western blot analysis was conducted to assess the protein levels. MTT and EdU assays were used to assess cell proliferation. Cell invasion was analyzed by transwell assay. Tube formation assay was conducted to detect cell angiogenesis. Dual-luciferase reporter assay and RNA immunoprecipitation were used to analyze the target relationship between miR-942-5p and circ_0015382 or NDRG1. Our data showed that circ_0015382 and NDRG1 were up-regulated, while miR-942-5p was down-regulated in the placental tissues of PE patients. Trophoblast cell proliferation, invasion, and angiogenesis were promoted by knockdown of circ_0015382. Circ_0015382 could sponge miR-942-5p, and NDRG1 was a target of miR-942-5p. MiR-942-5p inhibitor could reverse the promoting effects of si-circ_0015382 on trophoblast cell functions. Besides, the enhancing effects of miR-942-5p mimic on trophoblast cell proliferation, invasion, and angiogenesis could be eliminated by NDRG1 overexpression. In conclusion, our data showed that circ_0015382 inhibited trophoblast cell proliferation, invasion, and angiogenesis through regulating miR-942-5p/NDRG1 axis, providing a new mechanism for the regulation of PE progression.
Collapse
Affiliation(s)
- Haiyan Wu
- Obstetrical Department, Northwest Women's and Children's Hospital, No.1616 Yanxiang Road, Qujiang New District, Xi'an, Shaanxi, 710061, China
| | - Lijuan Zhao
- Obstetrical Department, Northwest Women's and Children's Hospital, No.1616 Yanxiang Road, Qujiang New District, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
15
|
Moura TDBD, Nunes FB, Crestani BDV, Araujo TFC, Hanauer EL, Corleta HVE, Branchini G. Preeclampsia and transport of ions and small molecules: A literature review. Placenta 2024; 156:77-91. [PMID: 39293185 DOI: 10.1016/j.placenta.2024.09.009] [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: 06/11/2024] [Revised: 08/22/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
Preeclampsia (PE) is a prevalent obstetric complication affecting approximately 3-5% of pregnancies worldwide and is a major cause of maternal and perinatal morbidity and mortality. Preeclampsia is considered a disease of the endothelial system that can progress to eclampsia, characterized by seizures. Early diagnosis and appropriate management are crucial to improving maternal and fetal outcomes, as preeclampsia can lead to severe complications such as placental abruption, fetal growth restriction, and stroke. The pathophysiology of PE is complex, involving a combination of genetic, acquired, and immunological factors. A central feature of the condition is inadequate placentation and impaired uteroplacental perfusion, leading to local hypoxia, endothelial dysfunction, vasoconstriction, and immunological dysregulation. Recent evidence suggests that dysregulation of ion transporters may play a significant role in the adaptation of uterine circulation during placentation. These transporters are essential for maintaining maternal-fetal homeostasis, influencing processes such as nutrient exchange, hormone synthesis, trophoblast cell migration, and the function of smooth muscle cells in blood vessels. In preeclampsia, adverse conditions like hypoxia and oxidative stress result in the downregulation of ion, solute, and water transporters, impairing their function. This review focuses on membrane transporters involved in PE, discussing functional alterations and their physiological implications. The goal of this investigation is to enhance understanding of how dysregulation of ion and small molecule transporters contributes to the development and progression of preeclampsia, underscoring the importance of exploring these signaling pathways for potential therapeutic interventions.
Collapse
Affiliation(s)
- Thaís Duarte Borges de Moura
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 245 Sarmento Leite St, Porto Alegre, RS, ZIP 90050170, Brazil
| | - Fernanda Bordignon Nunes
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 245 Sarmento Leite St, Porto Alegre, RS, ZIP 90050170, Brazil; Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), 6681 Ipiranga Av, Porto Alegre, RS, ZIP 90619-900, Brazil
| | - Bianca Dalla Vecchia Crestani
- Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 245 Sarmento Leite St, Porto Alegre, ZIP 90050170, Brazil
| | | | - Eduarda Luiza Hanauer
- Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 245 Sarmento Leite St, Porto Alegre, ZIP 90050170, Brazil
| | - Helena von Eye Corleta
- Departamento de Ginecologia e Obstetrícia, Faculdade de Medicina, Universidade Federal Do Rio Grande Do Sul (UFRGS), 2400 Ramiro Barcelos St, Porto Alegre, RS, ZIP 90035-003, Brazil
| | - Gisele Branchini
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 245 Sarmento Leite St, Porto Alegre, RS, ZIP 90050170, Brazil.
| |
Collapse
|
16
|
Jordan MM, Amabebe E, Khanipov K, Taylor BD. Scoping Review of Microbiota Dysbiosis and Risk of Preeclampsia. Am J Reprod Immunol 2024; 92:e70003. [PMID: 39440917 PMCID: PMC11501047 DOI: 10.1111/aji.70003] [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: 04/23/2024] [Revised: 08/20/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024] Open
Abstract
Limited studies have investigated the role of the microbiota in hypertensive disorders of pregnancy (HDP), particularly preeclampsia, which often results in preterm birth. We evaluated 23 studies that explored the relationship between gut, vaginal, oral, or placental microbiotas and HDP. Scopus, ProQuest Health Research Premium Collection, ProQuest Nursing & Allied Health Database, EBSCO, and Ovid were searched for relevant literature. Majority (18) of studies focused on the gut microbiota, and far fewer examined the oral cavity (3), vagina (3), and placenta (1). One study examined the gut, oral, and vaginal microbiotas. The consensus highlights a potential role for microbiota dysbiosis in preeclampsia and HDP. Especially in the third trimester, preeclampsia is associated with gut dysbiosis-deficient in beneficial species of Akkermansia, Bifidobacterium, and Coprococcus but enriched with pathogenic Campylobacterota and Candidatus Saccharibacteria, with low community α-diversity. Similarly, the preeclamptic vaginal and oral microbiotas are enriched with bacterial vaginosis and periodontal disease-associated species, respectively. The trend is also observed in the placenta, which is colonized by gastrointestinal, respiratory tract, and periodontitis-related pathogens. Consequently, a chronic proinflammatory state that adversely impacts placentation is implicated. These observations however require more mechanistic studies to establish the timing of the preceding immune dysfunction and any causality.
Collapse
Affiliation(s)
- Madeleine M. Jordan
- Division of Basic Science and Translational Research, University of Texas Medical Branch, Galveston, TX, USA
| | - Emmanuel Amabebe
- Division of Basic Science and Translational Research, University of Texas Medical Branch, Galveston, TX, USA
| | - Kamil Khanipov
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Brandie DePaoli Taylor
- Division of Basic Science and Translational Research, University of Texas Medical Branch, Galveston, TX, USA
- Department of Population Health and Health Disparities, School of Public and Population Health, Galveston, TX, USA
| |
Collapse
|
17
|
Khan MI, Khan MI, Wahab F. Irisin in Reproduction: Its Roles and Therapeutic Potential in Male and Female Fertility Disorders. Biomolecules 2024; 14:1222. [PMID: 39456155 PMCID: PMC11505643 DOI: 10.3390/biom14101222] [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: 05/20/2024] [Revised: 07/12/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
The current study focused on identifying the potential of irisin in mammalian reproduction. The established role of irisin, a proteolytic product of FNDC5, in adipose tissue browning, energy metabolism, and thermogenesis suggests its role in reproductive health, often disturbed by metabolic imbalances. Various studies on mice demonstrated irisin's role in improving spermatogenesis, sperm count, and testosterone levels by influencing the hypothalamus-pituitary-gonadal axis. Moreover, in females, there is a fluctuation in levels of irisin during critical reproductive stages, including menstrual cycles, puberty, and pregnancy. Conditions like pregnancy complications, precocious puberty, and polycystic ovary syndrome (PCOS) are found to have an association with abnormal irisin levels. The potential role of irisin in endometrial receptivity and preventing endometritis is also discussed in this review. Overall, the influence of irisin on female and male reproduction is evident from various studies. However, further research is needed to elucidate irisin mechanism in reproduction and its potential as a therapeutic or diagnostic tool for reproductive dysfunctions and infertility.
Collapse
Affiliation(s)
| | | | - Fazal Wahab
- Department of Biomedical Sciences, Pak-Austria Fachhochschule: Institute of Applied Sciences and Technology, Mang, Haripur 224000, Khyber Pakhtunkhwa, Pakistan; (M.I.K.)
| |
Collapse
|
18
|
Karakus S, Dogan HO. Exploring altered free amino acids and metabolites: Insights into the metabolic landscape of preeclampsia. Placenta 2024; 154:18-27. [PMID: 38850593 DOI: 10.1016/j.placenta.2024.06.002] [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: 12/28/2023] [Revised: 05/22/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
INTRODUCTION Preeclampsia (PE) is a complex disease that poses a risk for maternal and perinatal morbidity and mortality. This study aimed to investigate the role of maternal serum amino acids (AAs) levels in PE. MATERIALS AND METHODS A total of 56 pregnant women (26 with PE and 30 controls) were included in the study. The participants had a confirmed gestational age between 24 and 37 weeks. The mean body mass index (BMI) for the PE group was 33.1 kg/m2, while the control group had a mean BMI of 29.6 kg/m2. AAs levels were quantified, and statistical analyses were performed to identify significant differences between the two groups. Receiver Operating Characteristic (ROC) curve analysis was employed the diagnostic potential of specific AAs. RESULTS We observed significantly elevated levels of multiple AAs in the PE group, including citrulline, lysine, ethanolamine, ornithine and histidine. Citrulline exhibited exceptional predictive power for PE with 100.0% sensitivity and specificity at a cutoff of 7.79 µmol/L, reflected in an area under the curve (AUC) of 1.000. DISCUSSION Our study highlights the crucial involvement of altered amino acid levels, specifically in the urea cycle, disruptions in lysine and ethanolamine metabolism in PE development. Exploring these changes may reveal new therapeutic targets, providing insights into the disease's molecular mechanisms. Understanding amino acid metabolism in PE not only informs therapeutic strategies but also holds the potential to revolutionize early diagnosis and intervention.
Collapse
Affiliation(s)
- Savas Karakus
- Department of Obstetrics and Gynecology, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey.
| | - Halef Okan Dogan
- Department of Biochemistry, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey.
| |
Collapse
|
19
|
Sandberg M, Fomina T, Macsali F, Greve G, Øyen N, Leirgul E. Preeclampsia and neonatal outcomes in pregnancies with maternal congenital heart disease: A nationwide cohort study from Norway. Acta Obstet Gynecol Scand 2024; 103:1847-1858. [PMID: 38946266 PMCID: PMC11324925 DOI: 10.1111/aogs.14902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/30/2024] [Accepted: 06/10/2024] [Indexed: 07/02/2024]
Abstract
INTRODUCTION The prevalence of congenital heart disease (CHD) among women of reproductive age is rising. We aimed to investigate the risk of preeclampsia and adverse neonatal outcomes in pregnancies of mothers with CHD compared to pregnancies of mothers without heart disease. MATERIAL AND METHODS In a nationwide cohort of pregnancies in Norway 1994-2014, we retrieved information on maternal heart disease, the course of pregnancy, and neonatal outcomes from national registries. Comparing pregnancies with maternal CHD to pregnancies without maternal heart disease, we used Cox regression to estimate the adjusted hazard ratio (aHR) for preeclampsia and log-binomial regression to estimate the adjusted risk ratio (aRR) for adverse neonatal outcomes. The estimates were adjusted for maternal age and year of childbirth and presented with 95% confidence intervals (CIs). RESULTS Among 1 218 452 pregnancies, 2425 had mild maternal CHD, and 603 had moderate/severe CHD. Compared to pregnancies without maternal heart disease, the risk of preeclampsia was increased in pregnancies with mild and moderate/severe maternal CHD (aHR1.37, 95% CI 1.14-1.65 and aHR 1.62, 95% CI 1.13-2.32). The risk of preterm birth was increased in pregnancies with mild maternal CHD (aRR 1.33, 95% CI 1.15-1.54) and further increased with moderate/severe CHD (aRR 2.49, 95% CI 2.03-3.07). Maternal CHD was associated with elevated risks of both spontaneous and iatrogenic preterm birth. The risk of infants small-for-gestational-age was slightly increased with mild maternal CHD (aRR 1.12, 95% CI 1.00-1.26) and increased with moderate/severe CHD (aRR 1.63, 95% CI 1.36-1.95). The prevalence of stillbirth was 3.9 per 1000 pregnancies without maternal heart disease, 5.6 per 1000 with mild maternal CHD, and 6.8 per 1000 with moderate/severe maternal CHD. Still, there were too few cases to report a significant difference. There were no maternal deaths in women with CHD. CONCLUSIONS Moderate/severe maternal CHD in pregnancy was associated with increased risks of preeclampsia, preterm birth, and infants small-for-gestational-age. Mild maternal CHD was associated with less increased risks. For women with moderate/severe CHD, their risk of preeclampsia and adverse neonatal outcomes should be evaluated together with their cardiac risk in pregnancy, and follow-up in pregnancy should be ascertained.
Collapse
Affiliation(s)
- Marit Sandberg
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Tatiana Fomina
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Ferenc Macsali
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
- Norwegian Institute of Public Health, Oslo, Norway
| | - Gottfried Greve
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Nina Øyen
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Elisabeth Leirgul
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
20
|
Fato BR, de Alwis N, Beard S, Binder NK, Pritchard N, Kaitu'u-Lino TJ, Bubb KJ, Hannan NJ. Exploring the Therapeutic Potential of C-Type Natriuretic Peptide for Preeclampsia. Hypertension 2024; 81:1883-1894. [PMID: 39016006 DOI: 10.1161/hypertensionaha.124.22820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/21/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND Preeclampsia is a serious condition of pregnancy, complicated by aberrant maternal vascular dysfunction. CNP (C-type natriuretic peptide) contributes to vascular homeostasis, acting through NPR-B (natriuretic peptide receptor-B) and NPR-C (natriuretic peptide receptor-C). CNP mitigates vascular dysfunction of arteries in nonpregnant cohorts; this study investigates whether CNP can dilate maternal arteries in ex vivo preeclampsia models. METHODS Human omental arteries were dissected from fat biopsies collected during cesarean section. CNP, NPR-B, and NPR-C mRNA expression was assessed in arteries collected from pregnancies complicated by preeclampsia (n=6) and normotensive controls (n=11). Using wire myography, we investigated the effects of CNP on dilation of arteries from normotensive pregnancies. Arteries were preconstricted with either serum from patients with preeclampsia (n=6) or recombinant ET-1 (endothelin-1; vasoconstrictor elevated in preeclampsia; n=6) to model vasoconstriction associated with preeclampsia. Preconstricted arteries were treated with recombinant CNP (0.001-100 µmol/L) or vehicle and vascular relaxation assessed. In further studies, arteries were preincubated with NPR-B (5 µmol/L) and NPR-C (10 µmol/L) antagonists before serum-induced constriction (n=4-5) to explore mechanistic signaling. RESULTS CNP, NPR-B, and NPR-C mRNAs were not differentially expressed in omental arteries from preeclamptic pregnancies. CNP potently stimulated maternal artery vasorelaxation in our model of preeclampsia (using preeclamptic serum). Its vasodilatory actions were driven through the activation of NPR-B predominantly; antagonism of this receptor alone dampened CNP vasorelaxation. Interestingly, CNP did not reduce ET-1-driven omental artery constriction. CONCLUSIONS Collectively, these data suggest that enhancing CNP signaling through NPR-B offers a potential therapeutic strategy to reduce systemic vascular constriction in preeclampsia.
Collapse
Affiliation(s)
- Bianca R Fato
- Therapeutics Discovery and Vascular Function in Pregnancy Group (B.R.F., N.d.A., S.B., N.K.B., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Natasha de Alwis
- Therapeutics Discovery and Vascular Function in Pregnancy Group (B.R.F., N.d.A., S.B., N.K.B., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Sally Beard
- Therapeutics Discovery and Vascular Function in Pregnancy Group (B.R.F., N.d.A., S.B., N.K.B., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Natalie K Binder
- Therapeutics Discovery and Vascular Function in Pregnancy Group (B.R.F., N.d.A., S.B., N.K.B., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Natasha Pritchard
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Kristen J Bubb
- Department of Physiology, Biomedicine Discovery Institute (K.J.B.), Monash University, Clayton, Victoria, Australia
- Victorian Heart Institute, Faculty of Medicine, Nursing and Health Sciences (K.J.B.), Monash University, Clayton, Victoria, Australia
| | - Natalie J Hannan
- Therapeutics Discovery and Vascular Function in Pregnancy Group (B.R.F., N.d.A., S.B., N.K.B., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| |
Collapse
|
21
|
Soundararajan R, Khan T, Dadelszen PV. Pre-eclampsia challenges and care in low and middle-income countries: Understanding diagnosis, management, and health impacts in remote and developing regions. Best Pract Res Clin Obstet Gynaecol 2024; 96:102525. [PMID: 38964990 DOI: 10.1016/j.bpobgyn.2024.102525] [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: 11/21/2023] [Revised: 01/20/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
As an example of a low- and middle-income country (LMIC), India ranks pre-eclampsia among the top three causes of maternal mortality, following haemorrhage and infections. It is one of the primary concerns for maternal and perinatal health in LMICs. Many LMICs lack clear consensus and guidelines for the prevention, diagnosis, and management of hypertensive disorders in pregnancy, including pre-eclampsia. The International Society for the Study of Hypertension in Pregnancy 2021 guidelines address LMIC applications, offering customisable solutions. Atypical presentations of pre-eclampsia contribute to diagnostic delays, resulting in additional adverse maternal and perinatal outcomes. Implementing management strategies faces challenges in both urban and rural settings. Adapting global research involving local populations is imperative, with the potential for cost-effective adoption of international guidelines. Prevention, early diagnosis, and education dissemination are essential, involving healthcare providers and advocacy initiatives. Encouraging government investment in pre-eclampsia management as a public health initiative is important. This article explores socio-economic, cultural, and legislative factors influencing the management of pre-eclampsia in LMICs, addressing emerging challenges and potential partnerships for healthcare provision.
Collapse
Affiliation(s)
- Revathi Soundararajan
- Chief Consultant [Maternal Fetal Medicine], Managing Director, Mirror Health, Secretary, SMFM (I), Co-Chair, PEN (I), Bengaluru, India.
| | - Tamkin Khan
- Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, India.
| | | |
Collapse
|
22
|
Mayrink J, Miele MJ, Souza RT, Guida JP, Nobrega GM, Galvão RB, Costa ML, Fernandes KG, Capetini VC, Arantes AC, Anhê GF, Costa JL, Cecatti JG. Are vitamin D intake and serum levels in the mid-trimester of pregnancy associated with preeclampsia? Results from a Brazilian multicentre cohort. Pregnancy Hypertens 2024; 37:101150. [PMID: 39146694 DOI: 10.1016/j.preghy.2024.101150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/07/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024]
Abstract
OBJECTIVE To explore the association between serum levels and food intake of Vitamin D (VD) among healthy women in mid-pregnancy and preeclampsia. STUDY DESIGN In a Brazilian multicentre cohort of healthy nulliparous pregnant women from five maternity centres we developed a nested case-control analysis comparing cases with and without preeclampsia. Women were enrolled and followed during prenatal care, including only singleton pregnancies, without any fetal malformations or previous chronic maternal disease. We matched 87 cases of preeclampsia to eligible controls randomly selected in a 1:1 ratio, by age and region. MAIN OUTCOME MEASURES Blood samples from these were collected, and a 24-hour recall of food intake was obtained in mid-pregnancy, between 19 and 21 weeks. VD serum levels (25-hydroxyvitamin D) were measured by liquid chromatography-tandem mass spectrometry and were categorized as deficient, insufficient, and sufficient. The dietary intake of VD was estimated with the 24-hour diet recall applied at the same time and from supplementation. Maternal characteristics and VD levels were compared between cases and controls with OR and respective 95 %CI. Multivariate analysis using the Path method was used to assess relationships among VD, PE, BMI, skin colour/ethnicity, and diet. RESULTS The maternal characteristics of both groups were similar, except for the higher occurrence of obesity among women with preeclampsia (OR 3.47, 95 %CI 1.48-8.65). Dietary intake of VD was similar in both groups, and most of the women in both groups consumed insufficient VD (82.2 vs 79.3 % in the groups with and without PE). CONCLUSIONS Levels and dietary intake of VD were not associated with PE in this Brazilian sample of healthy pregnant women; however, BMI and skin colour/ethnicity were associated with PE.
Collapse
Affiliation(s)
- Jussara Mayrink
- Department of Obstetrics and Gynaecology, School of Medical Sciences, The University of Campinas, Campinas, SP, Brazil; Department of Obstetrics and Gynaecology, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Maria J Miele
- Department of Obstetrics and Gynaecology, School of Medical Sciences, The University of Campinas, Campinas, SP, Brazil
| | - Renato T Souza
- Department of Obstetrics and Gynaecology, School of Medical Sciences, The University of Campinas, Campinas, SP, Brazil
| | - Jose P Guida
- Department of Obstetrics and Gynaecology, School of Medical Sciences, The University of Campinas, Campinas, SP, Brazil
| | - Guilherme M Nobrega
- Department of Obstetrics and Gynaecology, School of Medical Sciences, The University of Campinas, Campinas, SP, Brazil
| | - Rafael B Galvão
- Department of Obstetrics and Gynaecology, School of Medical Sciences, The University of Campinas, Campinas, SP, Brazil
| | - Maria L Costa
- Department of Obstetrics and Gynaecology, School of Medical Sciences, The University of Campinas, Campinas, SP, Brazil
| | - Karayna G Fernandes
- Department of Obstetrics and Gynaecology, School of Medical Sciences, The University of Campinas, Campinas, SP, Brazil; Department of Obstetrics and Gynaecology, Jundiaí School of Medicine, Jundiaí, SP, Brazil
| | - Vinícius C Capetini
- Department of Translational Medicine, School of Medical Sciences, The University of Campinas, Campinas, SP, Brazil
| | - Ana Cf Arantes
- School of Pharmaceutical Sciences, the University of Campinas, Campinas, SP, Brazil
| | - Gabriel F Anhê
- Department of Translational Medicine, School of Medical Sciences, The University of Campinas, Campinas, SP, Brazil
| | - José L Costa
- School of Pharmaceutical Sciences, the University of Campinas, Campinas, SP, Brazil
| | - Jose G Cecatti
- Department of Obstetrics and Gynaecology, School of Medical Sciences, The University of Campinas, Campinas, SP, Brazil.
| |
Collapse
|
23
|
Rolnik DL, Syngelaki A, O'Gorman N, Wright D, Nicolaides KH, Poon LC. Aspirin for evidence-based preeclampsia prevention trial: effects of aspirin on maternal serum pregnancy-associated plasma protein A and placental growth factor trajectories in pregnancy. Am J Obstet Gynecol 2024; 231:342.e1-342.e9. [PMID: 38151219 DOI: 10.1016/j.ajog.2023.12.031] [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: 10/14/2023] [Revised: 12/14/2023] [Accepted: 12/20/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND The exact mechanism by which aspirin prevents preeclampsia remains unclear. Its effects on serum placental biomarkers throughout pregnancy are also unknown. OBJECTIVE To investigate the effects of aspirin on serum pregnancy-associated plasma protein A and placental growth factor trajectories using repeated measures from women at increased risk of preterm preeclampsia. STUDY DESIGN This was a longitudinal secondary analysis of the Combined Multimarker Screening and Randomized Patient Treatment with Aspirin for Evidence-based Preeclampsia Prevention trial using repeated measures of pregnancy-associated plasma protein A and placental growth factor. In the trial, 1620 women at increased risk of preterm preeclampsia were identified using the Fetal Medicine Foundation algorithm at 11 to 13+6 weeks of gestation, of whom 798 were randomly assigned to receive aspirin 150 mg and 822 to receive placebo daily from before 14 weeks to 36 weeks of gestation. Serum biomarkers were measured at baseline and follow-up visits at 19 to 24, 32 to 34, and 36 weeks of gestation. Generalized additive mixed models with treatment by gestational age interaction terms were used to investigate the effect of aspirin on biomarker trajectories over time. RESULTS Overall, there were 5507 pregnancy-associated plasma protein A and 5523 placental growth factor measurements. Raw pregnancy-associated plasma protein A values increased over time, and raw placental growth factor increased until 32 weeks of gestation followed by a decline. The multiple of the median mean values of the same biomarkers were consistently below 1.0 multiple of the median, reflecting the high-risk profile of the study population. Trajectories of mean pregnancy-associated plasma protein A and placental growth factor multiple of the median values did not differ significantly between the aspirin and placebo groups (aspirin treatment by gestational age interaction P values: .259 and .335, respectively). CONCLUSION In women at increased risk of preterm preeclampsia, aspirin 150 mg daily had no significant effects on pregnancy-associated plasma protein A or placental growth factor trajectories when compared to placebo.
Collapse
Affiliation(s)
- Daniel L Rolnik
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia.
| | - Argyro Syngelaki
- Fetal Medicine Research Institute, King's College Hospital, London, United Kingdom
| | - Neil O'Gorman
- Coombe Women and Infants University Hospital, Dublin, Ireland
| | - David Wright
- Institute of Health Research, University of Exeter, Exeter, United Kingdom
| | - Kypros H Nicolaides
- Fetal Medicine Research Institute, King's College Hospital, London, United Kingdom
| | - Liona C Poon
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong SAR
| |
Collapse
|
24
|
Stephansson O, Sandström A. Can short- and long-term maternal and infant risks linked to hypertension and diabetes during pregnancy be reduced by therapy? J Intern Med 2024; 296:216-233. [PMID: 39045893 DOI: 10.1111/joim.13823] [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] [Indexed: 07/25/2024]
Abstract
Hypertensive disorders of pregnancy (HDP), especially preeclampsia, and diabetes during pregnancy pose significant risks for both maternal and infant health, extending to long-term outcomes such as early-onset cardiovascular disease and metabolic disorders. Current strategies for managing HDP focus on screening, prevention, surveillance, and timely intervention. No disease-modifying therapies exist so far for established preeclampsia; delivery remains the definitive resolution. Preventive measures-including early pregnancy screening, exercise, and low-dose aspirin-show promise. Antihypertensive treatments reduce severe hypertension risks, whereas magnesium sulfate remains the standard for preventing eclampsia. Planned delivery from gestational week 37 can balance maternal benefits and neonatal risks in women with established preeclampsia. Delivery between 34 and 37 weeks gestation in women with preeclampsia has to balance risks for mother and infant. Lifestyle interventions-particularly diet and physical activity-are pivotal in managing gestational diabetes mellitus and type 2 diabetes. The oral antidiabetic metformin has shown benefits in glycaemic control and reducing maternal weight gain, although its long-term effects on offspring remain uncertain. The safety of other peroral antidiabetics in pregnancy is less studied. Advancements in glucose monitoring and insulin administration present encouraging prospects for enhancing outcomes in women with diabetes types 1 and 2. Both HDP and diabetes during pregnancy necessitate vigilant management through a combination of lifestyle modifications, pharmacological interventions, and timely obstetric care. Although certain treatments such as low-dose aspirin and metformin show efficacy in risk reduction, further research is ongoing to ensure safety for both mothers and their offspring to reduce short- and long-term adverse effects.
Collapse
Affiliation(s)
- Olof Stephansson
- Department of Medicine, Solna, Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
- Department of Women's Health, Division of Obstetrics, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Sandström
- Department of Medicine, Solna, Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
- Department of Women's Health, Division of Obstetrics, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
25
|
Adomi M, McElrath TF, Hernández-Díaz S, Vine SM, Huybrechts KF. TNF-α inhibitor use during pregnancy and the risk of preeclampsia: population-based cohort study. J Hypertens 2024; 42:1529-1537. [PMID: 38690936 PMCID: PMC11293998 DOI: 10.1097/hjh.0000000000003747] [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] [Indexed: 05/03/2024]
Abstract
BACKGROUND Although the clinical importance of preeclampsia is widely recognized, few treatment options are available for prevention. TNF-α inhibitors have been hypothesized to potentially prevent the disease. We aimed to examine whether exposure to TNF-α inhibitors during pregnancy reduces the risk of preeclampsia. METHODS We conducted a population-based pregnancy cohort study using nationwide samples of publicly (Medicaid data, 2000-2018) and commercially (MarketScan Research Database, 2003-2020) insured pregnant women linked to their liveborn infants. Exposure was ascertained based on a filled prescription or administration code for TNF-α inhibitors during the first and second trimester of pregnancy. The outcomes included early-onset preeclampsia, late-onset preeclampsia, and small-for-gestational age. For baseline confounding adjustment, we leveraged propensity score overlap weights to estimate risk ratios (RR). RESULTS Among 4 315 658 pregnancies in the Medicaid and the MarketScan cohort, 2736 (0.063%) were exposed to TNF-α inhibitors during the first trimester and 1712 (0.040%) during the second trimester. After adjustment, the risk of early-onset preeclampsia was not decreased among mothers exposed during the first trimester compared with unexposed women with treatment indications [RR pooled : 1.25, 95% confidence interval (CI) 0.93-1.67]. Similarly, the risk of late-onset preeclampsia was not decreased among mothers exposed during the second trimester compared with unexposed women (RR pooled : 0.99, 95% CI 0.81-1.22). CONCLUSION Contrary to the hypothesis, exposure to TNF-α inhibitors during pregnancy did not appear to be associated with a reduced risk of early-onset or late-onset preeclampsia. These findings do not support consideration of the use of TNF-α inhibitors for the prevention of preeclampsia.
Collapse
Affiliation(s)
- Motohiko Adomi
- Department of Epidemiology, Harvard T.H. Chan School of Public Health
| | - Thomas F McElrath
- Department of Epidemiology, Harvard T.H. Chan School of Public Health
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Brigham and Women's Hospital
| | | | - Seanna M Vine
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Krista F Huybrechts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
26
|
Manoharan MM, Montes GC, Acquarone M, Swan KF, Pridjian GC, Nogueira Alencar AK, Bayer CL. Metabolic theory of preeclampsia: implications for maternal cardiovascular health. Am J Physiol Heart Circ Physiol 2024; 327:H582-H597. [PMID: 38968164 PMCID: PMC11442029 DOI: 10.1152/ajpheart.00170.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Preeclampsia (PE) is a multisystemic disorder of pregnancy that not only causes perinatal mortality and morbidity but also has a long-term toll on the maternal and fetal cardiovascular system. Women diagnosed with PE are at greater risk for the subsequent development of hypertension, ischemic heart disease, cardiomyopathy, cerebral edema, seizures, and end-stage renal disease. Although PE is considered heterogeneous, inefficient extravillous trophoblast (EVT) migration leading to deficient spiral artery remodeling and increased uteroplacental vascular resistance is the likely initiation of the disease. The principal pathophysiology is placental hypoxia, causing subsequent oxidative stress, leading to mitochondrial dysfunction, mitophagy, and immunological imbalance. The damage imposed on the placenta in turn results in the "stress response" categorized by the dysfunctional release of vasoactive components including oxidative stressors, proinflammatory factors, and cytokines into the maternal circulation. These bioactive factors have deleterious effects on systemic endothelial cells and coagulation leading to generalized vascular dysfunction and hypercoagulability. A better understanding of these metabolic factors may lead to novel therapeutic approaches to prevent and treat this multisystemic disorder. In this review, we connect the hypoxic-oxidative stress and inflammation involved in the pathophysiology of PE to the resulting persistent cardiovascular complications in patients with preeclampsia.
Collapse
Affiliation(s)
- Mistina M Manoharan
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana, United States
| | - Guilherme C Montes
- Department of Pharmacology and Psychobiology, Roberto Alcântara Gomes Institute Biology (IBRAG), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Mariana Acquarone
- Department of Neurology, Tulane University, New Orleans, Louisiana, United States
| | - Kenneth F Swan
- Department of Obstetrics and Gynecology, Tulane University, New Orleans, Louisiana, United States
| | - Gabriella C Pridjian
- Department of Obstetrics and Gynecology, Tulane University, New Orleans, Louisiana, United States
| | | | - Carolyn L Bayer
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana, United States
- Department of Obstetrics and Gynecology, Tulane University, New Orleans, Louisiana, United States
| |
Collapse
|
27
|
Suksai M, Geater A, Amornchat P, Suntharasaj T, Suwanrath C, Pruksanusak N. Preeclampsia and timing of delivery: Disease severity, maternal and perinatal outcomes. Pregnancy Hypertens 2024; 37:101151. [PMID: 39208590 DOI: 10.1016/j.preghy.2024.101151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVES This study aimed to elucidate clinical characteristics, disease severity, and obstetric outcomes in women with pregnancy complicated with preeclampsia stratified by gestational age at delivery. STUDY DESIGN This retrospective study was conducted at a tertiary care facility from January 2011 to December 2020. MAIN OUTCOME MEASURES Maternal characteristics, risk factors, clinical signs and symptoms, laboratory test results, and maternal and perinatal outcomes were compared between early (<34 weeks) versus late (≥34 weeks) and preterm (<37 weeks) versus term (≥37 weeks) preeclampsia. RESULTS More than half of the women (56 %, 612/1094) had preterm preeclampsia. Overall, 30 % (329/1094) delivered before 34 weeks of gestation. Pregnancies with early preeclampsia had the worst maternal signs and symptoms, the highest median blood pressure level, and more abnormal laboratory abnormalities compared to those with late preeclampsia. Additionally, women with co-morbid diseases (chronic hypertension, chronic kidney disease, and systemic lupus erythematosus) were more likely to develop early than late preeclampsia. Of note, although adverse maternal and perinatal events occurred more commonly in early rather than late preeclampsia, 18 % (7/39) of eclampsia and 16 % (8/50) of hemolysis, elevated liver enzymes, and low platelet count syndrome cases occurred after 37 weeks of gestation. CONCLUSIONS Early preeclampsia posed the highest risk to the mother and infant(s); however, adverse maternal and perinatal events were still present even in cases of preeclampsia at term. Therefore, it is crucial for healthcare practitioners to remain vigilant and manage all cases with great care to prevent adverse outcomes.
Collapse
Affiliation(s)
- Manaphat Suksai
- Department of Obstetrics and Gynecology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand.
| | - Alan Geater
- Epidemiology Unit, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Pawinee Amornchat
- Department of Obstetrics and Gynecology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Thitima Suntharasaj
- Department of Obstetrics and Gynecology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Chitkasaem Suwanrath
- Department of Obstetrics and Gynecology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Ninlapa Pruksanusak
- Department of Obstetrics and Gynecology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| |
Collapse
|
28
|
Ramirez Zegarra R, Ghi T, Lees C. Does the use of angiogenic biomarkers for the management of preeclampsia and fetal growth restriction improve outcomes?: Challenging the current status quo. Eur J Obstet Gynecol Reprod Biol 2024; 300:268-277. [PMID: 39053087 DOI: 10.1016/j.ejogrb.2024.07.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
Monitoring and timing of delivery in preterm preeclampsia and fetal growth restriction is one of the biggest challenges in Obstetrics. Finding the optimal time of delivery of these fetuses usually involves a trade-off between the severity of the disease and prematurity. So far, most clinical guidelines recommend the use of a combination between clinical, laboratory and ultrasound markers to guide the time of delivery. Angiogenic biomarkers, especially placental growth factor (PlGF) and soluble fms-like tyrosine kinase-1 (sFlt-1), have gained significant attention in recent years for their potential role in the prediction and diagnosis of placenta-related disorders including preeclampsia and fetal growth restriction. Another potential clinical application of the angiogenic biomarkers is for the differential diagnosis of patients with chronic kidney disease, as this condition shares similar clinical features with preeclampsia. Consequently, angiogenic biomarkers have been advocated as tools for monitoring and deciding the optimal time of the delivery of fetuses affected by placental dysfunction. In this clinical opinion, we critically review the available literature on PlGF and sFlt-1 for the surveillance and time of the delivery in fetuses affected by preterm preeclampsia and fetal growth restriction. Moreover, we explore the use of angiogenic biomarkers for the differentiation between chronic kidney disease and superimposed preeclampsia.
Collapse
Affiliation(s)
- Ruben Ramirez Zegarra
- Department of Medicine and Surgery, Obstetrics and Gynecology Unit, University of Parma, Parma, Italy
| | - Tullio Ghi
- Department of Medicine and Surgery, Obstetrics and Gynecology Unit, University of Parma, Parma, Italy
| | - Christoph Lees
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom; Centre for Fetal Care, Queen Charlotte's and Chelsea Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom; Department of Development and Regeneration, Katholieke Universiteit Leuven, Leuven, Belgium
| |
Collapse
|
29
|
Ramdin S, Naicker T, Baijnath S, Govender N. Is renal dysfunction amplified in an arginine vasopressin induced rat model of preeclampsia? Reprod Biol 2024; 24:100910. [PMID: 38851025 DOI: 10.1016/j.repbio.2024.100910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 04/27/2024] [Accepted: 05/25/2024] [Indexed: 06/10/2024]
Abstract
Renal dysfunction is important in preeclampsia (PE) pathophysiology and has not been fully explored in the arginine vasopressin (AVP) rat model of PE. This study aimed to determine kidney toxicity associated with this model. Female Sprague Dawley rats (n = 24) were subcutaneously infused with AVP or saline for 18 days. Urine samples (GD8, 14 and 18) were used to determine the levels of albumin, VEGF-A, clusterin, NGAL/Lipocalin-2, KIM-1, cystatin C, TIMP-1, β2M and OPN via Multiplex ELISAs. Albumin, and NGAL/lipocalin-2 were significantly elevated in the PAVP vs PS group on GD14 and GD18 (p < 0.001) respectively. VEGF-A significantly decreased in the pregnant vs non-pregnant groups on GD14 and 18 (p < 0.001). Clusterin (p < 0.001) and OPN (p < 0.05) were significantly higher in the PAVP vs PS group on GD18. Cystatin C and KIM-1 are significantly upregulated in the PAVP vs PS groups throughout gestation (p < 0.05). β2M is significantly elevated in the PAVP vs PS group on GD14 and 18 (p < 0.05). AVP elevated the urinary levels of the kidney injury biomarkers and replicated the renal dysfunction associated with PE development. Our findings confirm the potential applications of this model in studying the mechanisms underlying renal damage in PE.
Collapse
Affiliation(s)
- Sapna Ramdin
- Department of Basic Medical Sciences, Faculty of Health Sciences, Durban University of Technology, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Sooraj Baijnath
- Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nalini Govender
- Department of Basic Medical Sciences, Faculty of Health Sciences, Durban University of Technology, Durban, South Africa.
| |
Collapse
|
30
|
Zhao H, Wen P, Xiong Y, Xu Q, Zi Y, Zheng X, Chen S, Qin Y, Shao S, Tu X, Zheng Z, Li X. Association of sleep traits with risk of hypertensive disorders of pregnancy: a mendelian randomization study. J Hypertens 2024; 42:1606-1614. [PMID: 38780189 PMCID: PMC11296273 DOI: 10.1097/hjh.0000000000003771] [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: 02/28/2024] [Revised: 04/21/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Unhealthy sleep patterns are common during pregnancy and have been associated with an increased risk of developing hypertensive disorders of pregnancy (HDPs) in observational studies. However, the causality underlying these associations remains uncertain. This study aimed to evaluate the potential causal association between seven sleep traits and the risk of HDPs using a two-sample Mendelian randomization study. METHODS Genome-wide association study (GWAS) summary statistics were obtained from the FinnGen consortium, UK Biobank, and other prominent consortia, with a focus on individuals of European ancestry. The primary analysis utilized an inverse-variance-weighted MR approach supplemented by sensitivity analyses to mitigate potential biases introduced by pleiotropy. Furthermore, a two-step MR framework was employed for mediation analyses. RESULTS The data analyzed included 200 000-500 000 individuals for each sleep trait, along with approximately 15 000 cases of HDPs. Genetically predicted excessive daytime sleepiness (EDS) exhibited a significant association with an increased risk of HDPs [odds ratio (OR) 2.96, 95% confidence interval (95% CI) 1.40-6.26], and the specific subtype of preeclampsia/eclampsia (OR 2.97, 95% CI 1.06-8.3). Similarly, genetically predicted obstructive sleep apnea (OSA) was associated with a higher risk of HDPs (OR 1.27, 95% CI 1.09-1.47). Sensitivity analysis validated the robustness of these associations. Mediation analysis showed that BMI mediated approximately 25% of the association between EDS and HDPs, while mediating up to approximately 60% of the association between OSA and the outcomes. No statistically significant associations were observed between other genetically predicted sleep traits, such as chronotype, daytime napping, sleep duration, insomnia, snoring, and the risk of HDPs. CONCLUSION Our findings suggest a causal association between two sleep disorders, EDS and OSA, and the risk of HDPs, with BMI acting as a crucial mediator. EDS and OSA demonstrate promise as potentially preventable risk factors for HDPs, and targeting BMI may represent an alternative treatment strategy to mitigate the adverse impact of sleep disorders.
Collapse
Affiliation(s)
- Huanqiang Zhao
- Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong
| | - Ping Wen
- Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong
| | - Yu Xiong
- Obstetrics and Gynecology Hospital, Fudan University
- The Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Qixin Xu
- Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong
| | - Yang Zi
- Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong
| | - Xiujie Zheng
- Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong
| | - Shiguo Chen
- Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong
| | - Yueyuan Qin
- Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong
| | - Shuyi Shao
- Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong
| | - Xinzhi Tu
- Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong
| | - Zheng Zheng
- Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong
| | - Xiaotian Li
- Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong
- Obstetrics and Gynecology Hospital, Fudan University
- The Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| |
Collapse
|
31
|
Chatzakis C, Papavasiliou D, Mansukhani T, Nicolaides KH, Charakida M. Maternal vascular-placental axis in the third trimester in women with gestational diabetes mellitus, hypertensive disorders, and unaffected pregnancies. Am J Obstet Gynecol 2024:S0002-9378(24)00900-1. [PMID: 39218286 DOI: 10.1016/j.ajog.2024.08.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 08/10/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Hypertensive disorders of pregnancy and gestational diabetes mellitus are characterized by vascular dysfunction and are associated with long term cardiovascular risks. OBJECTIVE This study aimed to compare different markers of maternal vascular function in women with gestational diabetes mellitus, preeclampsia, or gestational hypertension and in women whose pregnancies were unaffected by these complications and to assess the association between maternal vascular function and markers of placental perfusion and maternal vascular-placental axis in 4 groups of women. STUDY DESIGN This was a prospective observational study of women who had routine hospital visits at 35 0/7 to 36 6/7 weeks of gestation at King's College Hospital, London, United Kingdom. The routine hospital visit included recording of maternal demographic characteristics and medical history, ultrasound examination for fetal anatomy and growth, Doppler studies of the uterine arteries and ophthalmic arteries, carotid-femoral pulse wave velocity measurements, estimation of the augmentation index and total peripheral resistance, and measurements of serum placental growth factor and soluble fms-like tyrosine kinase 1. Linear regression analysis was performed for the outcomes of uterine artery pulsatility index multiple of the median, placental growth factor multiple of the median, and soluble fms-like tyrosine kinase 1 multiple of the median. The ophthalmic artery peak systolic velocity ratio, pulse wave velocity, augmentation index, and total peripheral vascular resistance were assessed as potential predictors. This analysis was performed on all women and separately in the different groups. RESULTS The study population of 6502 women included 614 (9.4%) with gestational diabetes mellitus, 140 (2.1%) who subsequently developed preeclampsia, and 129 (2.0%) who developed gestational hypertension. Women with gestational diabetes mellitus had increased pulse wave velocity compared with those with pregnancies unaffected by gestational diabetes mellitus, preeclampsia, or gestational hypertension. Women with preeclampsia or gestational hypertension had lower placental growth factor multiple of the median and higher uterine artery pulsatility index multiple of the median, soluble fms-like tyrosine kinase 1 multiple of the median, augmentation index, pulse wave velocity, total peripheral resistance, and ophthalmic artery peak systolic velocity ratio than those with unaffected pregnancies. In women with unaffected pregnancies, the ophthalmic artery peak systolic velocity ratio was predictive of the uterine artery pulsatility index multiple of the median, and ophthalmic artery peak systolic velocity ratio, augmentation index, total peripheral resistance, and pulse wave velocity were predictive of the placental growth factor multiple of the median and the soluble fms-like tyrosine kinase 1 multiple of the median. In women with gestational diabetes mellitus, the ophthalmic artery peak systolic velocity ratio was predictive of the uterine artery pulsatility index multiple of the median; the ophthalmic artery peak systolic velocity ratio, total peripheral resistance, and pulse wave velocity were predictive of the placental growth factor multiple of the median; and total peripheral resistance was predictive of the soluble fms-like tyrosine kinase 1 multiple of the median. In women with preeclampsia, the ophthalmic artery peak systolic velocity ratio was predictive of the uterine artery pulsatility index multiple of the median, placental growth factor multiple of the median, and soluble fms-like tyrosine kinase 1 multiple of the median. In women unaffected by gestational diabetes mellitus, preeclampsia, or gestational hypertension, the ophthalmic artery peak systolic velocity ratio was predictive of the uterine artery pulsatility index multiple of the median, and the augmentation index, total peripheral resistance, pulse wave velocity, and the ophthalmic artery peak systolic velocity ratio were predictive of the placental growth factor multiple of the median and the soluble fms-like tyrosine kinase 1 multiple of the median. CONCLUSION In the third trimester of pregnancy, women with preeclampsia, gestational hypertension, and gestational diabetes mellitus present with increased arterial stiffness. In addition, women diagnosed with hypertensive complications showed increased peripheral vascular resistance. The ophthalmic artery peak systolic velocity ratio provided predictive information for placental perfusion and function in all pregnant women, whereas vascular indices were more informative for placental function in women with unaffected pregnancies and those with gestational diabetes mellitus than in those with preeclampsia or gestational hypertension. Our data suggest that vascular assessment in women during pregnancy not only may provide information about maternal vascular health but also can be used to provide information about individual risk factors for placental insufficiency. The selection of the vascular index will have to be tailored according to the maternal profile and pregnancy complication.
Collapse
Affiliation(s)
- Christos Chatzakis
- Second Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - Dimitra Papavasiliou
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom
| | - Tanvi Mansukhani
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom
| | - Kypros H Nicolaides
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom.
| | - Marietta Charakida
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom; School of Biomedical Engineering and Imaging Sciences, King's College Hospital, London, United Kingdom
| |
Collapse
|
32
|
Huang Z, Sun L, Gao Y, Shi M, Zhang P, Ding Y, Wang J, Wei J, Yang X, Li R. Exploration of the molecular characteristics and potential clinical significance of shared immune-related genes between preterm preeclampsia and term preeclampsia. BMC Pregnancy Childbirth 2024; 24:543. [PMID: 39148025 PMCID: PMC11328443 DOI: 10.1186/s12884-024-06526-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 04/16/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Preeclampsia is a severe obstetric disorder that significantly affects the maternal and neonatal peri-partum safety and long-term quality of life. However, there is limited research exploring the common mechanisms and potential clinical significance between early-onset preeclampsia and full-term preeclampsia from an immunological perspective. METHODS In this study, data analysis was conducted. Initially, immune-related co-expressed genes involving both subtypes of preeclampsia were identified through Weighted Gene Co-expression Network Analysis (WGCNA). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were further employed to investigate the shared pathways regulated by immune-related genes. Binary logistic regression identified co-expressed genes with diagnostic value for preeclampsia, and a diagnostic model was constructed. Gene Set Enrichment Analysis (GSEA) predicted the potential biological functions of the selected genes. Lasso and Cox regression analyses identified genes closely associated with gestational duration, and a risk score model was established. A 4-gene feature, immune-related gene model for predicting the risk of preterm birth in preeclamptic pregnant women, was developed and validated through qPCR experiments. Immune cell infiltration analysis determined differences in immune cell infiltration between the two subtypes of preeclampsia. RESULTS This study identified 4 immune-related co-expressed genes (CXCR6, PIK3CB, IL1RAP, and OSMR). Additionally, diagnostic and preterm birth risk prediction models for preeclampsia were constructed based on these genes. GSEA analysis suggested the involvement of these genes in the regulation of galactose metabolism, notch signaling pathway, and RIG-I like receptor signaling pathway. Immune pathway analysis indicated that the activation of T cell co-inhibition could be a potential intervention target for immunotherapy in early-onset preeclampsia. CONCLUSION Our study provides promising insights into immunotherapy and mechanistic research for preeclampsia, discovering novel diagnostic and intervention biomarkers, and offering personalized diagnostic tools for preeclampsia.
Collapse
Affiliation(s)
- Zhengrui Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Road west, Tianhe district, Guangdong, 510630, China
| | - Lu Sun
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Road west, Tianhe district, Guangdong, 510630, China
| | - Yudie Gao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Road west, Tianhe district, Guangdong, 510630, China
| | - Meiting Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Road west, Tianhe district, Guangdong, 510630, China
| | - Ping Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Road west, Tianhe district, Guangdong, 510630, China
| | - Yuzhen Ding
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, 518000, China
| | - Jian Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Road west, Tianhe district, Guangdong, 510630, China
| | - Jiachun Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Road west, Tianhe district, Guangdong, 510630, China
| | - Xiuli Yang
- Department of Obstetrics and Gynecology, The Sixth Affiliated Hospital of Jinan University, No. 88, Changdong Road, Changping town, Dongguan, Guangdong, 523000, China.
| | - Ruiman Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Road west, Tianhe district, Guangdong, 510630, China.
| |
Collapse
|
33
|
Rezende KBDC, Barmpas DS, Werner H, Rolnik DL, Costa FDS, Poon L, Pedroso MA, Bernardes LS, Rezende JC, Ragazini CS, Bornia RG, Wright D, Nicolaides K, Pritsivelis C, de Sá RAM. Comment on: Prediction and secondary prevention of preeclampsia from the perspective of public health management - the initiative of the State of Rio de Janeiro. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRÍCIA 2024; 46:e-rbgo80. [PMID: 39176200 PMCID: PMC11341189 DOI: 10.61622/rbgo/2024rbgo80] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 06/06/2024] [Indexed: 08/24/2024] Open
Affiliation(s)
- Karina Bilda de Castro Rezende
- Universidade Federal do Rio de JaneiroMaternity SchoolRio de JaneiroRJBrazilMaternity School, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Danielle Sodré Barmpas
- Fundação Oswaldo CruzInstituto Fernandes FigueiraRio de JaneiroRJBrazilInstituto Fernandes Figueira, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil.
| | - Heron Werner
- Pontifícia Universidade Católica do Rio de JaneiroDepartment of Fetal MedicineRio de JaneiroRJBrazilDepartment of Fetal Medicine, Pontifícia Universidade Católica do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Daniel Lorber Rolnik
- Monash UniversityDepartment of Obstetrics and GynaecologyMelbourneAustraliaDepartment of Obstetrics and Gynaecology, Monash University, Melbourne, Australia.
| | - Fabricio da Silva Costa
- Griffith UniversityGold Coast University Hospital and School of Medicine and DentistryMaternal Fetal Medicine UnitGold CoastAustraliaMaternal Fetal Medicine Unit, Gold Coast University Hospital and School of Medicine and Dentistry, Griffith University, Gold Coast, Australia.
| | - Liona Poon
- The Chinese University of Hong KongDepartment of Obstetrics and GynecologyHong KongChinaDepartment of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong.
| | - Marianna Amaral Pedroso
- Maternal Fetal Medicine UnitBelo HorizonteMGBrazilMaternal Fetal Medicine Unit, Oncoclínicas, Belo Horizonte, MG, Brazil.
| | - Lisandra Stein Bernardes
- Aalborg UniversityNorth Denmark Regional HospitalObstetrics and Gynecology DepartmentDenmarkCentre for Clinical Research, Obstetrics and Gynecology Department, North Denmark Regional Hospital, Aalborg University, Denmark.
| | - Juliana Costa Rezende
- Universidade de BrasíliaDepartment of Obstetrics and GynecologyBrasíliaDFBrazilDepartment of Obstetrics and Gynecology, Universidade de Brasília, Brasília, DF, Brazil.
| | - Conrado Sávio Ragazini
- Universidade de São PauloFaculdade de MedicinaRibeirão PretoSPBrazilFaculdade de Medicina, Universidade de São Paulo, Ribeirão Preto, SP, Brazil.
| | - Rita Guérios Bornia
- Universidade Federal do Rio de JaneiroMaternity SchoolRio de JaneiroRJBrazilMaternity School, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - David Wright
- University of ExeterInstitute of Health ResearchExeterUKInstitute of Health Research, University of Exeter, Exeter, UK.
| | - Kypros Nicolaides
- King's College HospitalFetal Medicine Research InstituteLondonUKFetal Medicine Research Institute, King's College Hospital, London, UK.
| | - Cristos Pritsivelis
- Universidade Federal do Rio de JaneiroMaternity SchoolRio de JaneiroRJBrazilMaternity School, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Renato Augusto Moreira de Sá
- Fundação Oswaldo CruzInstituto Fernandes FigueiraRio de JaneiroRJBrazilInstituto Fernandes Figueira, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil.
- Universidade Federal de FluminenseDepartment of Obstetrics and GynaecologyRio de JaneiroRJBrazilDepartment of Obstetrics and Gynaecology, Universidade Federal de Fluminense, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
34
|
Patel E, Suresh S, Mueller A, Bisson C, Zhu K, Verlohren S, Dadelszen PV, Magee L, Rana S. sFlt1/PlGF among patients with suspected preeclampsia when considering hypertensive status. AJOG GLOBAL REPORTS 2024; 4:100359. [PMID: 39005612 PMCID: PMC11239699 DOI: 10.1016/j.xagr.2024.100359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND In high-resource settings, biomarkers of angiogenic balance, such as the soluble fms-like tyrosine kinase-1 (sFlt1)/placental growth factor (PlGF) ratio, have been studied extensively to aid in evaluation of patients with suspected preeclampsia (PE), and have been incorporated into the 2021 International Society for the Study of Hypertension in Pregnancy definition of PE. The utility in under-resourced settings has not been as well characterized. OBJECTIVE This analysis sought to identify the role of the sFlt1/PlGF ratio in the evaluation of patients with or without hypertension who are suspected of having PE without other diagnostic information. STUDY DESIGN This is a secondary analysis of a prior prospective study of patients who were presented with suspected PE at ≥20+0 weeks' gestation at a single academic tertiary care center. Patients were recruited in the parent study from July 2009 to June 2012. In the original study, clinicians were masked to biomarker results, and patients were followed by chart review. In this analysis, the performance of the sFlt1/PlGF ratio (≤38, >38, or >85) was assessed alone in identifying both hypertensive and non-hypertensive patients at risk of evolving into PE with severe features (PE-SF; American College of Obstetricians and Gynecologists' definition) within two weeks of the triage visit (PE-SF2). Hypertension was defined as a blood pressure (BP)≥140/90 mmHg. RESULTS There were 1043 patients included in the analysis; of whom, 579 (55.5%) and 464 (44.5%) presented with or without hypertension, respectively. In triage, 332 (75.4%) of hypertensive patients presented due to BP concerns, and the remainder were evaluated due to other features (new-onset headache, proteinuria, or edema). On triage evaluation, 66.8% of all patients had a normal sFlt1/PlGF ratio ≤38, and 17.0% had an elevated ratio >85. Among hypertensive patients, a sFlt1/PlGF ratio ≤38 was a good rule-out test for PE-SF2 (negative likelihood ratio [LR-] of 0.15), and a ratio >85 was a good rule-in test (positive likelihood ratio [LR+] of 5.75). Among normotensive patients, sFlt1/PlGF was useful as a rule-in test for ratio >38 (LR+ 5.13) and >85 (LR+ 12.80). Stratified by gestational age, sFlt1/PlGF continued to be a good rule in and good rule out test at <35 weeks among those with hypertension but did not have good test performance ≥35 weeks. sFlt1/PlGF had a good test performance as a rule in test for >85 regardless of gestational age. In triage, 4.3% (30/693) of patients with sFlt1/PlGF ratio <38 had concurrent laboratory evidence of PE, compared with 15.9% (28/176) patients with ratio >85. CONCLUSION These findings support the potential for the use of sFlt1/PlGF and BP measurement alone in resource-limited settings where other laboratory tests or clinical expertise are unavailable for risk stratification. Performance of the biomarker varied by the presence of hypertension and gestational age.
Collapse
Affiliation(s)
- Easha Patel
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago Medicine, Chicago, IL (Dr Patel, Ms Mueller, Drs Bisson, Zhu, Rana)
| | - Sunitha Suresh
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, NorthShore University Health System, Evanston, IL (Dr Suresh)
| | - Ariel Mueller
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago Medicine, Chicago, IL (Dr Patel, Ms Mueller, Drs Bisson, Zhu, Rana)
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA (Ms Mueller)
| | - Courtney Bisson
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago Medicine, Chicago, IL (Dr Patel, Ms Mueller, Drs Bisson, Zhu, Rana)
| | - Katherine Zhu
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago Medicine, Chicago, IL (Dr Patel, Ms Mueller, Drs Bisson, Zhu, Rana)
| | - Stefan Verlohren
- Charité - Universitätsmedizin Berlin, Berlin, Germany (Dr Verlohren)
| | - Peter Von Dadelszen
- Institute of Women and Children's Health, Department of Women and Children's Health, School of Life Course and Population Sciences, King's College London, London, United Kingdom (Drs Dadelszen, Magee)
| | - Laura Magee
- Institute of Women and Children's Health, Department of Women and Children's Health, School of Life Course and Population Sciences, King's College London, London, United Kingdom (Drs Dadelszen, Magee)
| | - Sarosh Rana
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago Medicine, Chicago, IL (Dr Patel, Ms Mueller, Drs Bisson, Zhu, Rana)
| |
Collapse
|
35
|
Handmark M, Lin A, Edsfeldt A, Sarno G, Fraser A, Rich-Edwards JW, Gonҫalves I, Pihlsgård M, Timpka S. STEMI, Revascularization, and Peak Troponin by Adverse Pregnancy Outcomes in Women With Myocardial Infarction. JACC. ADVANCES 2024; 3:101088. [PMID: 39070091 PMCID: PMC11277779 DOI: 10.1016/j.jacadv.2024.101088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/15/2024] [Accepted: 05/07/2024] [Indexed: 07/30/2024]
Abstract
Background Women with a history of adverse pregnancy outcomes have a higher risk of coronary heart disease. Emerging evidence suggests that women with a history of preeclampsia have a different pattern of overall coronary atherosclerosis and that they at the time of myocardial infarction (MI) more frequently present with ST-segment elevation MI (STEMI) compared to women with no such history. Objectives The purpose of this study was to determine whether among women with MI, those with a history of adverse pregnancy outcomes are more likely to present with STEMI or other clinical characteristics indicating a more severe myocardial injury. Methods The study sample consisted of 8,320 women aged ≤65 years with first MI in Sweden 2007 to 2022. Regression models were used to estimate the association between adverse pregnancy outcomes (hypertensive disorders of pregnancy [non-preeclamptic hypertension and preeclampsia], small for gestational age [SGA] infant, and preterm delivery) and STEMI, invasive revascularization, and high troponin, while considering known predictors of coronary heart disease. Results In total, 3,128 (38%) of women suffered STEMI. The adjusted OR of presenting with STEMI were higher in women with a history of preterm preeclampsia (OR: 1.40; 95% CI: 1.05-1.88), or an SGA infant (OR: 1.30; 95% CI: 1.13-1.50) compared to women with no such history, as well as for in-hospital revascularization. Stratified by infarct type, troponin levels did not differ by adverse pregnancy outcome history. Conclusions Among women with a first MI, a history of preterm preeclampsia or SGA infant were associated with STEMI and invasive revascularization.
Collapse
Affiliation(s)
- Moa Handmark
- Perinatal and Cardiovascular Epidemiology and Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Annie Lin
- Perinatal and Cardiovascular Epidemiology and Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Andreas Edsfeldt
- Cardiovascular Research - Translational Studies, Lund University, Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden
| | - Giovanna Sarno
- Department of Medical Sciences, Cardiology and Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Abigail Fraser
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Janet W. Rich-Edwards
- Division of Women’s Health, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Isabel Gonҫalves
- Cardiovascular Research - Translational Studies, Lund University, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden
| | - Mats Pihlsgård
- Perinatal and Cardiovascular Epidemiology and Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Simon Timpka
- Perinatal and Cardiovascular Epidemiology and Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Department of Obstetrics and Gynaecology, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
36
|
Conley MK. Preeclampsia: Short- and Long-Term Effects. Neonatal Netw 2024; 43:234-246. [PMID: 39164098 DOI: 10.1891/nn-2024-0001] [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] [Indexed: 08/22/2024]
Abstract
Does our time inside the womb predict our future? Evidence suggests that the environment in the womb plays a powerful role in predicting specific adult diseases. The fetus is constantly responding and adapting to the intrauterine environment by a process called programming. Toxic exposures, such as nutritional deficits and hypoxia, can affect fetal development and increase the risk for specific diseases that manifest later in our adult life. Preeclampsia (PE) is one disorder that results in a less-than-optimal environment for the growing fetus. It is pregnancy-specific and defined as new-onset hypertension after 20 weeks' gestation in the presence of maternal multiorgan dysfunction. To the best of our understanding, the pathogenesis is multifactorial and involves dysfunction of the placenta and the vascular, renal, and immunological systems. Treatment options are limited and may result in adverse outcomes for the fetus and newborn. Preeclampsia is a major contributor to perinatal and maternal morbidity and mortality worldwide, thus generating a significant healthcare burden. Research continues to demonstrate that mothers and infants affected by PE are at increased susceptibility to chronic conditions such as cardiovascular, renal, metabolic, and neurological diseases. More efforts are needed to further understand this disease. Efforts to increase awareness will help improve clinical outcomes for both mothers and infants.
Collapse
|
37
|
Mammadova N, Özler S, Özdemir BG, Avcı F, Koçak N, Çintesun E, Örgül G, Çelik Ç. The role of the Chitinase 3-Like 1 (CHI3L1) genes in the preeclampsia pathophysiology. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20231574. [PMID: 39045955 PMCID: PMC11288277 DOI: 10.1590/1806-9282.20231574] [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: 01/13/2024] [Accepted: 03/10/2024] [Indexed: 07/25/2024]
Abstract
OBJECTIVE The aim of this study was to investigate the relationship between Chitinase 3-Like 1 gene polymorphisms and the occurrence of preeclampsia in a selected cohort of pregnant women. METHODS A total of 75 pregnant women participated in the study, 35 of whom were diagnosed with preeclampsia, while 40 served as healthy controls. The preeclamptic group was subdivided based on severity. Real-time polymerase chain reaction was employed to analyze the serum samples for variations in Chitinase 3-Like 1 gene polymorphisms. RESULTS The rs880633 polymorphism was found to be significantly more frequent in the control group (80%) compared with the overall preeclamptic group (60%) (p<0.05). In the severity-based subgroups, rs880633 appeared in 57.1% of non-severe and 61.9% of severe preeclamptics. Contrarily, the heterozygous form of rs7515776 polymorphism showed a significantly higher prevalence in the preeclamptic cohort (p<0.05), without distinctions in severity subgroups. CONCLUSION The study suggests that the rs880633 polymorphism may serve a protective role against the development of preeclampsia, whereas the rs7515776 polymorphism may be associated with an elevated risk. Further research is warranted to elucidate the clinical implications of these findings.
Collapse
Affiliation(s)
- Nigar Mammadova
- Afiyet Hospital, Private Clinic of Obstetrics and Gynecology – İstanbul, Turkey
| | - Sibel Özler
- Selcuk University, Faculty of Medicine, Department of Obstetrics and Gynecology – Konya, Turkey
| | - Belma Gözde Özdemir
- Selcuk University, Faculty of Medicine, Department of Obstetrics and Gynecology – Konya, Turkey
| | - Fazıl Avcı
- Selcuk University, Faculty of Medicine, Department of Obstetrics and Gynecology – Konya, Turkey
| | - Nadir Koçak
- Selcuk University, Faculty of Medicine, Department of Medical Genetics – Konya, Turkey
| | - Ersin Çintesun
- Selcuk University, Faculty of Medicine, Department of Obstetrics and Gynecology – Konya, Turkey
| | - Gökçen Örgül
- Selcuk University, Faculty of Medicine, Department of Obstetrics and Gynecology – Konya, Turkey
| | - Çetin Çelik
- Selcuk University, Faculty of Medicine, Department of Obstetrics and Gynecology – Konya, Turkey
| |
Collapse
|
38
|
Campos MAA, Oppermann MLR, Sanseverino MTV, Guerra GL, Hirakata VN, Reichelt AJ. Congenital anomalies in pregnancies with overt and pregestational type 2 diabetes: a gray portrayal from a cohort in Brazil. Diabetol Metab Syndr 2024; 16:157. [PMID: 38992793 PMCID: PMC11238503 DOI: 10.1186/s13098-024-01376-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/10/2024] [Indexed: 07/13/2024] Open
Abstract
OBJECTIVE To describe the frequency and types of congenital anomalies and associated risk factors in Brazilian women with type 2 diabetes. METHODS In this retrospective cohort study between 2005 and 2021, we included all pregnant participants with type 2 diabetes from the two major public hospitals in southern Brazil. We collected data from the electronic hospital records. Congenital anomalies were classified by the 10th revised International Classification of Diseases, Q chapter, enhanced by the EUROCAT registry classification, and categorized by type and gravity. We used multiple Poisson regression with robust estimates to estimate risks. RESULTS Among 648 participants, we excluded 19, and 62 were lost to follow-up; therefore, we included 567 participants. Overt diabetes arose in 191 participants (33.7%, 95% CI 30.0% - 38.0%). Less than 20% of the participants supplemented folate. Congenital anomalies occurred in 78 neonates (13.8%, CI 11.0 - 16.9%), 73 babies (93.6%) presented major anomalies, and 20 (10.5%) cases occurred in participants with overt diabetes. Cardiac anomalies were the most frequent (43 isolated and 12 combined). Pre-eclampsia was associated with an increased risk in the analyses including all women (adjusted RR 1.87 (95% CI 1.23-2.85), p = 0.003), but not in analyses including only women with an HbA1c measured up to the 14th gestational age. HbA1c, either measured at any time in pregnancy (adjusted RR 1.21 (95% CI 1.10-1.33), p < 0.001) or up to the first 14 weeks (adjusted RR 1.22, 95% CI 1.10-1.35, p < 0.001) was the only sustained risk factor. Risk factors such as maternal age, obesity, diabetes diagnosis, or use of antidiabetic medications were not associated with congenital anomalies. CONCLUSION We found a high frequency of congenital anomalies associated with poor maternal glycemic control and revealed an almost universal lack of preconception care. An urgent call to action is mandatory for the reversal of this gray scenario.
Collapse
Affiliation(s)
- Maria Amélia A Campos
- Serviço de Endocrinologia, Hospital Nossa Senhora da Conceição, Porto Alegre, Brazil
| | - Maria Lúcia R Oppermann
- Serviço de Ginecologia e Obstetrícia, Hospital de Clínicas de Porto Alegre, and Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Maria Teresa V Sanseverino
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, and Faculdade de Medicina da Pontifícia Universidade Católica, Porto Alegre, Brazil
| | - Giulia L Guerra
- Faculdade de Medicina, Universidade Feevale, Novo Hamburgo, Brazil
| | - Vânia N Hirakata
- Unidade de Bioestatística e Análise de Dados, Hospital de Clínicas de Porto Alegre, and Programa de Pós-graduação em Cardiologia e Ciências Cardiovasculares, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Angela J Reichelt
- Serviço de Endocrinologia e Metabologia, Hospital de Clínicas de Porto Alegre, and Programa de Pós‑Graduação em Ciências Médicas: Endocrinologia, Universidade Federal do Rio Grande do Sul Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
39
|
Torres-Torres J, Espino-y-Sosa S, Martinez-Portilla R, Borboa-Olivares H, Estrada-Gutierrez G, Acevedo-Gallegos S, Ruiz-Ramirez E, Velasco-Espin M, Cerda-Flores P, Ramirez-Gonzalez A, Rojas-Zepeda L. A Narrative Review on the Pathophysiology of Preeclampsia. Int J Mol Sci 2024; 25:7569. [PMID: 39062815 PMCID: PMC11277207 DOI: 10.3390/ijms25147569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Preeclampsia (PE) is a multifactorial pregnancy disorder characterized by hypertension and proteinuria, posing significant risks to both maternal and fetal health. Despite extensive research, its complex pathophysiology remains incompletely understood. This narrative review aims to elucidate the intricate mechanisms contributing to PE, focusing on abnormal placentation, maternal systemic response, oxidative stress, inflammation, and genetic and epigenetic factors. This review synthesizes findings from recent studies, clinical trials, and meta-analyses, highlighting key molecular and cellular pathways involved in PE. The review integrates data on oxidative stress biomarkers, angiogenic factors, immune interactions, and mitochondrial dysfunction. PE is initiated by poor placentation due to inadequate trophoblast invasion and improper spiral artery remodeling, leading to placental hypoxia. This triggers the release of anti-angiogenic factors such as soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin (sEng), causing widespread endothelial dysfunction and systemic inflammation. Oxidative stress, mitochondrial abnormalities, and immune dysregulation further exacerbate the condition. Genetic and epigenetic modifications, including polymorphisms in the Fms-like tyrosine kinase 1 (FLT1) gene and altered microRNA (miRNA) expression, play critical roles. Emerging therapeutic strategies targeting oxidative stress, inflammation, angiogenesis, and specific molecular pathways like the heme oxygenase-1/carbon monoxide (HO-1/CO) and cystathionine gamma-lyase/hydrogen sulfide (CSE/H2S) pathways show promise in mitigating preeclampsia's effects. PE is a complex disorder with multifactorial origins involving abnormal placentation, endothelial dysfunction, systemic inflammation, and oxidative stress. Despite advances in understanding its pathophysiology, effective prevention and treatment strategies remain limited. Continued research is essential to develop targeted therapies that can improve outcomes for both mothers and their babies.
Collapse
Affiliation(s)
- Johnatan Torres-Torres
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Salvador Espino-y-Sosa
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Raigam Martinez-Portilla
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Hector Borboa-Olivares
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Guadalupe Estrada-Gutierrez
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Sandra Acevedo-Gallegos
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Erika Ruiz-Ramirez
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Martha Velasco-Espin
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Pablo Cerda-Flores
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Andrea Ramirez-Gonzalez
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Lourdes Rojas-Zepeda
- Maternal-Fetal Medicine Department, Instituto Materno Infantil del Estado de Mexico, Toluca 50170, Mexico
| |
Collapse
|
40
|
Cuenca-Gómez D, De Paco Matallana C, Rolle V, Mendoza M, Valiño N, Revello R, Adiego B, Casanova MC, Molina FS, Delgado JL, Wright A, Figueras F, Nicolaides KH, Santacruz B, Gil MM. Comparison of different methods of first-trimester screening for preterm pre-eclampsia: cohort study. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2024; 64:57-64. [PMID: 38411276 DOI: 10.1002/uog.27622] [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: 02/01/2024] [Revised: 02/10/2024] [Accepted: 02/13/2024] [Indexed: 02/28/2024]
Abstract
OBJECTIVE To compare the predictive performance of three different mathematical models for first-trimester screening of pre-eclampsia (PE), which combine maternal risk factors with mean arterial pressure (MAP), uterine artery pulsatility index (UtA-PI) and serum placental growth factor (PlGF), and two risk-scoring systems. METHODS This was a prospective cohort study performed in eight fetal medicine units in five different regions of Spain between September 2017 and December 2019. All pregnant women with singleton pregnancy and a non-malformed live fetus attending their routine ultrasound examination at 11 + 0 to 13 + 6 weeks' gestation were invited to participate in the study. Maternal characteristics and medical history were recorded and measurements of MAP, UtA-PI, serum PlGF and pregnancy-associated plasma protein-A (PAPP-A) were converted into multiples of the median (MoM). Risks for term PE, preterm PE (< 37 weeks' gestation) and early PE (< 34 weeks' gestation) were calculated according to the FMF competing-risks model, the Crovetto et al. logistic regression model and the Serra et al. Gaussian model. PE classification was also performed based on the recommendations of the National Institute for Health and Care Excellence (NICE) and the American College of Obstetricians and Gynecologists (ACOG). We estimated detection rates (DR) with their 95% CIs at a fixed 10% screen-positive rate (SPR), as well as the area under the receiver-operating-characteristics curve (AUC) for preterm PE, early PE and all PE for the three mathematical models. For the scoring systems, we calculated DR and SPR. Risk calibration was also assessed. RESULTS The study population comprised 10 110 singleton pregnancies, including 32 (0.3%) that developed early PE, 72 (0.7%) that developed preterm PE and 230 (2.3%) with any PE. At a fixed 10% SPR, the FMF, Crovetto et al. and Serra et al. models detected 82.7% (95% CI, 69.6-95.8%), 73.8% (95% CI, 58.7-88.9%) and 79.8% (95% CI, 66.1-93.5%) of early PE; 72.7% (95% CI, 62.9-82.6%), 69.2% (95% CI, 58.8-79.6%) and 74.1% (95% CI, 64.2-83.9%) of preterm PE; and 55.1% (95% CI, 48.8-61.4%), 47.1% (95% CI, 40.6-53.5%) and 53.9% (95% CI, 47.4-60.4%) of all PE, respectively. The best correlation between predicted and observed cases was achieved by the FMF model, with an AUC of 0.911 (95% CI, 0.879-0.943), a slope of 0.983 (95% CI, 0.846-1.120) and an intercept of 0.154 (95% CI, -0.091 to 0.397). The NICE criteria identified 46.7% (95% CI, 35.3-58.0%) of preterm PE at 11% SPR and ACOG criteria identified 65.9% (95% CI, 55.4-76.4%) of preterm PE at 33.8% SPR. CONCLUSIONS The best performance of screening for preterm PE is achieved by mathematical models that combine maternal factors with MAP, UtA-PI and PlGF, as compared to risk-scoring systems such as those of NICE and ACOG. While all three algorithms show similar results in terms of overall prediction, the FMF model showed the best performance at an individual level. © 2024 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- D Cuenca-Gómez
- Department of Obstetrics and Gynecology, Hospital Universitario de Torrejón, Torrejón de Ardoz, Madrid, Spain
- Faculty of Medicine, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - C De Paco Matallana
- Department of Obstetrics and Gynecology, Hospital Clínico Universitario Virgen de la Arrixaca, El Palmar, Murcia, Spain
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, El Palmar, Murcia, Spain
| | - V Rolle
- Biostatistics and Clinical Research Unit, Hospital Universitario de Torrejón, Torrejón de Ardoz, Madrid, Spain
| | - M Mendoza
- Department of Obstetrics and Gynecology, Hospital Universitari Vall d'Hebrón, Barcelona, Catalonia, Spain
| | - N Valiño
- Department of Obstetrics and Gynecology, Complejo Hospitalario Universitario A Coruña, A Coruña, Galicia, Spain
| | - R Revello
- Department of Obstetrics and Gynecology, Hospital Universitario Quirón, Pozuelo de Alarcón, Madrid, Spain
| | - B Adiego
- Department of Obstetrics and Gynecology, Hospital Universitario Fundación de Alcorcón, Alcorcón, Madrid, Spain
| | - M C Casanova
- Department of Obstetrics and Gynecology, Hospital Universitario de Torrejón, Torrejón de Ardoz, Madrid, Spain
- Faculty of Medicine, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - F S Molina
- Department of Obstetrics and Gynecology, Hospital Universitario San Cecilio, Granada, Spain
- Instituto de Investigación Biosanitaria Ibs, Granada, Spain
| | - J L Delgado
- Department of Obstetrics and Gynecology, Hospital Clínico Universitario Virgen de la Arrixaca, El Palmar, Murcia, Spain
| | - A Wright
- Institute of Health Research, University of Exeter, Exeter, UK
| | - F Figueras
- BCNatal-Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital San Joan de Deu, Barcelona, Spain
| | - K H Nicolaides
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - B Santacruz
- Department of Obstetrics and Gynecology, Hospital Universitario de Torrejón, Torrejón de Ardoz, Madrid, Spain
- Faculty of Medicine, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - M M Gil
- Department of Obstetrics and Gynecology, Hospital Universitario de Torrejón, Torrejón de Ardoz, Madrid, Spain
- Faculty of Medicine, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| |
Collapse
|
41
|
Martín-Palumbo G, Alcorta MD, de Aguado MP, Antolín E, Bartha JL. Urinary sFlt-1 and PlGF as preeclampsia predictors: sFlt-1/creatinine ratio improves the prediction value. Eur J Obstet Gynecol Reprod Biol 2024; 298:53-60. [PMID: 38728842 DOI: 10.1016/j.ejogrb.2024.05.002] [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: 02/09/2024] [Revised: 04/28/2024] [Accepted: 05/04/2024] [Indexed: 05/12/2024]
Abstract
OBJECTIVES To evaluate the correlation between maternal serum and urinary soluble Fms-like tyrosine kinase-1 (sFlt-1) and placental growth factor (PlGF) levels and to assess their potential value in preeclampsia and fetal growth restriction. STUDY DESIGN This case-control longitudinal prospective study was performed in 49 singleton pregnant women, divided into two clinical groups, low risk pregnancy (n = 23) and pregnancy complicated by preeclampsia (n = 26). Maternal serum and urinary sFlt-1 and PlGF levels were quantified by electrochemiluminescence. Every patient underwent an ultrasound for fetal biometry. Doppler assessment was done when estimated fetal weight was under the 10th centile. ROC curves were used to evaluate the predictive capability of serum and urinary angiogenic biomarkers and their ratios on preeclampsia. Linear regression was used to compare the values of serum and urinary sFlt-1 and PlGF and their ratios. RESULTS Urine biomarkers were positively associated with their serum values, being the best associated urinary PlGF (R2 = 0.73), which also showed the highest predictive capability of preeclampsia of urine biomarkers (AUC 0.866). The predictive capability of urinary sFlt-1 was much lower (AUC 0.640), but increased when adjusting by serum creatinine, a more precise parameter (AUC 0.863). CONCLUSIONS Urinary PlGF could be a lesser invasive alternative to circulating biomarkers to monitor pregnancies complicated with preeclampsia that need repeated controls of their pregnancy complication. Urinary sFlt-1 values need adjustment by serum creatinine to be reliable.
Collapse
Affiliation(s)
- Giovanna Martín-Palumbo
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, La Paz University Hospital, Madrid, Spain.
| | | | - Marta Pérez de Aguado
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, La Paz University Hospital, Madrid, Spain
| | - Eugenia Antolín
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, La Paz University Hospital, Madrid, Spain
| | - José Luis Bartha
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, La Paz University Hospital, Madrid, Spain; Obstetrics and Gynecology of the Autonomous University of Madrid, Director of the Maternal and Fetal Research Group, Fundación para la Investigación Biomédica, La Paz University Hospital, Madrid, Spain
| |
Collapse
|
42
|
Thomopoulos C, Hitij JB, De Backer T, Gkaliagkousi E, Kreutz R, Lopez-Sublet M, Marketou M, Mihailidou AS, Olszanecka A, Pechère-Bertschi A, Pérez MP, Persu A, Piani F, Socrates T, Stolarz-Skrzypek K, Cífková R. Management of hypertensive disorders in pregnancy: a Position Statement of the European Society of Hypertension Working Group 'Hypertension in Women'. J Hypertens 2024; 42:1109-1132. [PMID: 38690949 DOI: 10.1097/hjh.0000000000003739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Hypertensive disorders in pregnancy (HDP), remain the leading cause of adverse maternal, fetal, and neonatal outcomes. Epidemiological factors, comorbidities, assisted reproduction techniques, placental disorders, and genetic predisposition determine the burden of the disease. The pathophysiological substrate and the clinical presentation of HDP are multifarious. The latter and the lack of well designed clinical trials in the field explain the absence of consensus on disease management among relevant international societies. Thus, the usual clinical management of HDP is largely empirical. The current position statement of the Working Group 'Hypertension in Women' of the European Society of Hypertension (ESH) aims to employ the current evidence for the management of HDP, discuss the recommendations made in the 2023 ESH guidelines for the management of hypertension, and shed light on controversial issues in the field to stimulate future research.
Collapse
Affiliation(s)
- Costas Thomopoulos
- Department of Cardiology, General Hospital of Athens 'Laiko', Athens, Greece
| | - Jana Brguljan Hitij
- Department of Hypertension, University Medical Centre Ljubljana, Medical University Ljubljana, Slovenia
| | - Tine De Backer
- Cardiovascular Center & Clinical Pharmacology, University Hospital Gent, Belgium
| | - Eugenia Gkaliagkousi
- 3rd Department of Internal Medicine, Papageorgiou Hospital Faculty of Medicine, Aristotle University of Thessaloniki, Greece
| | - Reinhold Kreutz
- Charite-Universitätsmedizin Berlin, Institute of Clinical Pharmacology and Toxicology, Berlin, Germany
| | - Marilucy Lopez-Sublet
- AP-HP, Hopital Avicenne, Centre d'Excellence Europeen en Hypertension Arterielle, Service de Medecine Interne, INSERM UMR 942 MASCOT, Paris 13-Universite Paris Nord, Bobigny, FCRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists)
| | - Maria Marketou
- School of Medicine, University of Crete, Heraklion, Greece
| | - Anastasia S Mihailidou
- Department of Cardiology and Kolling Institute, Royal North Shore Hospital, St Leonards
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Agnieszka Olszanecka
- 1st Department of Cardiology, Interventional Electrocardiology, and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| | | | - Mariana Paula Pérez
- Department of Hypertension. Hospital de Agudos J. M. Ramos Mejía, Buenos Aires, Argentina
| | - Alexandre Persu
- Division of Cardiology, Cliniques Universitaires Saint-Luc and Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Federica Piani
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Thenral Socrates
- Medical Outpatient and Hypertension Clinic, ESH Hypertension Centre of Excellence University Hospital Basel, Basel, Switzerland
| | - Katarzyna Stolarz-Skrzypek
- 1st Department of Cardiology, Interventional Electrocardiology, and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| | - Renata Cífková
- Center for Cardiovascular Prevention, Charles University in Prague, First Faculty of Medicine and Thomayer University Hospital
- Department of Medicine II, Charles University in Prague, First Faculty of Medicine, Prague, Czech Republic
| |
Collapse
|
43
|
Dimopoulou S, Neculcea D, Papastefanou I, Galan A, Androulaki M, Nicolaides KH, Charakida M. Impact of hypertensive disorders of pregnancy on offspring cardiovascular function: from fetal life to early childhood. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2024; 64:44-49. [PMID: 38437242 DOI: 10.1002/uog.27627] [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: 01/15/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/06/2024]
Abstract
OBJECTIVE Epidemiological studies suggest that, following in-utero exposure to hypertensive disorder of pregnancy (HDP), children may be at increased long-term cardiovascular risk, but data in early childhood are lacking. We aimed to investigate the independent influence of HDP on infant cardiac structure and function, after accounting for differences in childhood risk-factor profile. METHODS This was a longitudinal study of 71 children born of a pregnancy complicated by HDP (gestational hypertension or pre-eclampsia) and 304 children born of a normotensive pregnancy. Detailed cardiovascular assessment was performed at mid gestation and at a median of 2.3 (interquartile range, 2.1-2.4) years postnatally. Linear mixed-effects modeling was used to determine the independent influence of HDP on infant cardiac function and structure after accounting for differences in childhood risk-factor profile. RESULTS There were no differences in demographic characteristics between children whose mother developed HDP and those born of a normotensive pregnancy, but delivery was earlier and birth weight was lower in the HDP group. In fetal life, there were no significant differences in cardiac function or structure between the HDP and non-HDP groups. In early childhood, in the HDP group compared with the non-HDP group, there was greater relative wall thickness (mean ± SD, 0.7 ± 0.3 vs 0.6 ± 0.3; P = 0.047) and increased left ventricular mass (indexed to body surface area) (mean ± SD, 80.9 ± 20.4 g/m2 vs 75.7 ± 16.5 g/m2; P = 0.024); however, these differences did not persist on multivariable analysis. Longitudinal analysis revealed that there was no difference in the change in cardiac functional indices from fetal life to early childhood between the HDP and non-HDP groups. CONCLUSION There is no evidence that HDP has an adverse effect on offspring cardiovascular health in fetal life or in early childhood. © 2024 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- S Dimopoulou
- Harris Birthright Research Centre for Fetal Medicine, Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - D Neculcea
- Harris Birthright Research Centre for Fetal Medicine, Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - I Papastefanou
- Department of Women and Children's Health, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - A Galan
- Harris Birthright Research Centre for Fetal Medicine, Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - M Androulaki
- Harris Birthright Research Centre for Fetal Medicine, Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - K H Nicolaides
- Harris Birthright Research Centre for Fetal Medicine, Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - M Charakida
- Harris Birthright Research Centre for Fetal Medicine, Fetal Medicine Research Institute, King's College Hospital, London, UK
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| |
Collapse
|
44
|
Park SK, Kim H, Kim Y, Jang YE, Kim JT. Effect of epidural anesthesia on the optic nerve sheath diameter in patients with pre-eclampsia: a prospective observational study. Reg Anesth Pain Med 2024:rapm-2024-105444. [PMID: 38950931 DOI: 10.1136/rapm-2024-105444] [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: 03/04/2024] [Accepted: 06/18/2024] [Indexed: 07/03/2024]
Abstract
INTRODUCTION Optic nerve sheath diameter (ONSD) reflects intracranial pressure and is increased in pre-eclampsia. Administrating a significant volume of epidural solution into the epidural space can potentially increase ONSD. We investigated the impact of epidural local anesthetic injection on ONSD in patients with pre-eclampsia. METHODS Patients with pre-eclampsia (n=11) and normotensive pregnant women (n=11) received de novo epidural anesthesia for cesarean delivery. We administered 21 mL of an epidural solution containing 2% lidocaine and 50 μg fentanyl into the lumbar epidural space in incremental doses. ONSD was measured at baseline, 3, 10, and 20 min after completing the epidural injection, after delivery, and at the end of surgery. Primary outcome was the change in ONSD from baseline to 3 min after epidural injection in patients with pre-eclampsia and normotensive pregnant women. Serial changes in the ONSD were analyzed using a linear mixed model. RESULTS At baseline and 3 min after epidural drug injection, ONSD was significantly larger in patients with pre-eclampsia than in normotensive mothers (5.7 vs 4.1 mm, p=0.001 and 5.4 vs 4.1 mm, p<0.001, respectively). However, there were no significant changes in ONSD at 3 min after injection from baseline in either group (p>0.999). Linear mixed model demonstrated that ONSD did not change after epidural anesthesia in either group (p=0.279 and p=0.347, respectively). CONCLUSIONS Despite a higher baseline ONSD in pre-eclampsia, epidural anesthesia did not further increase ONSD. Our findings indicate that epidural anesthesia can be safely administered in patients with pre-eclampsia at risk of increased intracranial pressure, without other intracranial pathology. TRIAL REGISTRATION NUMBER NCT04095832.
Collapse
Affiliation(s)
- Sun-Kyung Park
- Department of Anesthesiology and Pain Medicine and Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hansol Kim
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Youngwon Kim
- Department of Anesthesiology and Pain Medicine and Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young-Eun Jang
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin-Tae Kim
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
45
|
Cooke WR, Jones GD, Redman CW, Vatish M. Small RNAs in the pathogenesis of preeclampsia. Placenta 2024:S0143-4004(24)00285-6. [PMID: 38955620 DOI: 10.1016/j.placenta.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/28/2024] [Accepted: 06/16/2024] [Indexed: 07/04/2024]
Abstract
Preeclampsia is a major contributor to maternal and fetal morbidity and mortality. The disorder can be classified into early- and late-onset subtypes, both of which evolve in two stages. The first stage comprises the development of pre-clinical, utero-placental malperfusion. Early and late utero-placental malperfusion have different causes and time courses. Early-onset preeclampsia (20 % of cases) is driven by dysfunctional placentation in the first half of pregnancy. In late-onset preeclampsia (80 % of cases), malperfusion is a consequence of placental compression within the confines of a limited uterine cavity. In both subtypes, the malperfused placenta releases stress signals into the maternal circulation. These stress signals trigger onset of the clinical syndrome (the second stage). Small RNA molecules, which are implicated in cellular stress responses in general, may be involved at different stages. Micro RNAs contribute to abnormal trophoblast invasion, immune dysregulation, angiogenic imbalance, and syncytiotrophoblast-derived extracellular vesicle signalling in preeclampsia. Transfer RNA fragments are placental signals known to be specifically involved in cell stress responses. Disorder-specific differences in small nucleolar RNAs and piwi-interacting RNAs have also been reported. Here, we summarise key small RNA advances in preeclampsia pathogenesis. We propose that existing small RNA classifications are unhelpful and that non-biased assessment of RNA expression, incorporation of non-annotated molecules and consideration of chemical modifications to RNAs may be important in elucidating preeclampsia pathogenesis.
Collapse
Affiliation(s)
- William R Cooke
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Level 3 Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| | - Gabriel Davis Jones
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Level 3 Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Christopher Wg Redman
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Level 3 Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Manu Vatish
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Level 3 Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| |
Collapse
|
46
|
Sabren S, Hagar T, Khateeb N, Evgeny F, Yara FN, Perlitz Y, Farid N. Placental and serum levels of human α-Klotho in preeclampsia & intra-uterine growth retardation: A potential sensitive biomarker? Pregnancy Hypertens 2024; 36:101115. [PMID: 38608394 DOI: 10.1016/j.preghy.2024.101115] [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: 01/22/2024] [Accepted: 02/17/2024] [Indexed: 04/14/2024]
Abstract
INTRODUCTION α-Klotho protein has three isoforms: a transmembrane (mKL), a shed- soluble isoform, and a circulating soluble isoform (sKL). mKL is expressed in the kidney and placenta, while sKL is detectable in blood and urine. It is known that α-Klotho levels fluctuate during pregnancy mainly in women with complications such as preeclampsia (PE) and intra-uterine growth restriction (IUGR). METHODS Forty-nine participants were divided into two groups: healthy and complicated pregnancy (PE, IUGR or both). Tissue samples (2 cm3) from the maternal side, Blood and urine samples were collected during pregnancy and postpartum. Samples were subjected to biochemical (WB), histological (H&E and IHC) staining as well as genetic analysis (qPCR). RESULTS Blood αKL levels were preserved in both healthy and complicated pregnancies. Significantly lower blood αKL concentrations were found in PE postpartum (PP) compared to levels during pregnancy, and were significantly lower compared with postpartum of a healthy pregnancy. αKL activity was reduced in complicated pregnancies vs. healthy pregnancies. Placen tal mKL levels (ELISA) and expression (WB) were lowered in complicated pregnancies compared with the healthy pregnancies group. Additionally, we found a significant decline in the expression of mKL mRNA in PE/IUGR placentas compared with the healthy group. DISCUSSION Several studies have focused on the involvement of αKL in normal placentation during pregnancy. Our results suggest lower function of sKL in complicated pregnancy compared with a control, and present differences in placental mKL levels as well as tissue and gene expression between healthy and complicated pregnancy. In light of our results, we conclude that complicated pregnancy is associated with in decline in mKL.
Collapse
Affiliation(s)
- Shehada Sabren
- Diabetes and Metabolism Lab, Tzafon Medical Center, Israel; Azrieli Faculty of Medicine, Safed, Bar Ilan University, Ramat-Gan, Israel
| | - Tadmor Hagar
- Diabetes and Metabolism Lab, Tzafon Medical Center, Israel
| | - Nardeen Khateeb
- Gynecological and Obstetrics Department, Tzafon Medical Center, Israel
| | | | - Francis-Nakhle Yara
- Gynecological and Obstetrics Department, Galilee Medical Center, Nahariya, Israel
| | - Youri Perlitz
- Gynecological and Obstetrics Department, Tzafon Medical Center, Israel
| | - Nakhoul Farid
- Azrieli Faculty of Medicine, Safed, Bar Ilan University, Ramat-Gan, Israel; Cardiovascular Laboratory, Medical Research Institute, The Galilee Medical Center, Nahariya, Israel.
| |
Collapse
|
47
|
Wilson MG, Bone JN, Slade LJ, Mistry HD, Singer J, Crozier SR, Godfrey KM, Baird J, von Dadelszen P, Magee LA. Blood pressure measurement and adverse pregnancy outcomes: A cohort study testing blood pressure variability and alternatives to 140/90 mmHg. BJOG 2024; 131:1006-1016. [PMID: 38054262 PMCID: PMC11256866 DOI: 10.1111/1471-0528.17724] [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: 07/25/2023] [Revised: 10/19/2023] [Accepted: 11/07/2023] [Indexed: 12/07/2023]
Abstract
OBJECTIVE To examine the association with adverse pregnancy outcomes of: (1) American College of Cardiology/American Heart Association blood pressure (BP) thresholds, and (2) visit-to-visit BP variability (BPV), adjusted for BP level. DESIGN An observational study. SETTING Analysis of data from the population-based UK Southampton Women's Survey (SWS). POPULATION OR SAMPLE 3003 SWS participants. METHODS Generalised estimating equations were used to estimate crude and adjusted relative risks (RRs) of adverse pregnancy outcomes by BP thresholds, and by BPV (as standard deviation [SD], average real variability [ARV] and variability independent of the mean [VIM]). Likelihood ratios (LRs) were calculated to evaluate diagnostic test properties, for BP at or above a threshold, compared with those below. MAIN OUTCOME MEASURES Gestational hypertension, severe hypertension, pre-eclampsia, preterm birth (PTB), small-for-gestational-age (SGA) infants, neonatal intensive care unit (NICU) admission. RESULTS A median of 11 BP measurements were included per participant. For BP at ≥20 weeks' gestation, higher BP was associated with more adverse pregnancy outcomes; however, only BP <140/90 mmHg was a good rule-out test (negative LR <0.20) for pre-eclampsia and BP ≥140/90 mmHg a good rule-in test (positive LR >8.00) for the condition. BP ≥160/110 mmHg could rule-in PTB, SGA infants and NICU admission (positive LR >5.0). Higher BPV (by SD, ARV, or VIM) was associated with gestational hypertension, severe hypertension, pre-eclampsia, PTB, SGA and NICU admission (adjusted RRs 1.05-1.39). CONCLUSIONS While our findings do not support lowering the BP threshold for pregnancy hypertension, they suggest BPV could be useful to identify elevated risk of adverse outcomes.
Collapse
Affiliation(s)
- Milly G. Wilson
- Department of Women and Children's Health, Faculty of Medicine, School of Life Course and Population SciencesKing's College LondonLondonUK
| | - Jeffrey N. Bone
- British Columbia Children's Hospital Research InstituteUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of Obstetrics and GynaecologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Laura J. Slade
- Robinson Research InstituteThe University of AdelaideAdelaideSouth AustraliaAustralia
- Department of Obstetrics and GynaecologyWomen's and Children's HospitalAdelaideSouth AustraliaAustralia
| | - Hiten D. Mistry
- Department of Women and Children's Health, Faculty of Medicine, School of Life Course and Population SciencesKing's College LondonLondonUK
| | - Joel Singer
- School of Population and Public HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Sarah R. Crozier
- MRC Lifecourse Epidemiology CentreUniversity of SouthamptonSouthamptonUK
- NIHR Applied Research Collaboration Wessex, Southampton Science ParkSouthamptonUK
| | - Keith M. Godfrey
- MRC Lifecourse Epidemiology CentreUniversity of SouthamptonSouthamptonUK
- NIHR Southampton Biomedical Research CentreUniversity of Southampton and University Hospital Southampton NHS Foundation TrustSouthamptonUK
| | - Janis Baird
- MRC Lifecourse Epidemiology CentreUniversity of SouthamptonSouthamptonUK
- NIHR Applied Research Collaboration Wessex, Southampton Science ParkSouthamptonUK
- NIHR Southampton Biomedical Research CentreUniversity of Southampton and University Hospital Southampton NHS Foundation TrustSouthamptonUK
| | - Peter von Dadelszen
- Department of Women and Children's Health, Faculty of Medicine, School of Life Course and Population SciencesKing's College LondonLondonUK
| | - Laura A. Magee
- Department of Women and Children's Health, Faculty of Medicine, School of Life Course and Population SciencesKing's College LondonLondonUK
| |
Collapse
|
48
|
Zeng C, Liu H, Wang Z, Li J. Novel insights into the complex interplay of immune dysregulation and inflammatory biomarkers in preeclampsia and fetal growth restriction: A two-step Mendelian randomization analysis. J Transl Autoimmun 2024; 8:100226. [PMID: 38225945 PMCID: PMC10788291 DOI: 10.1016/j.jtauto.2023.100226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 01/17/2024] Open
Abstract
Background The relationship between genetic immune dysregulation and the occurrence of preeclampsia (PE) or PE with fetal growth restriction (PE with FGR) has yielded inconsistent findings, and the underlying mediators of this association remain elusive. We aimed to explore the causal impact of genetic immune dysregulation on the risk of PE or PE with FGR and to elucidate the role of specific transcriptomes in mediating this relationship. Methods A two-step Mendelian randomization (MR) analysis was performed to explore the link between immune dysregulation and PE or PE with FGR, as well as to identify potential inflammatory biomarkers that act as mediators. GWAS summary data for outcomes were obtained from the FinnGen dataset. The analyses encompassed five systemic immune-associated diseases, four chronic genital inflammatory diseases, and thirty-one inflammatory biomarkers. Summary-data-based MR (SMR) and HEIDI analysis were conducted to test whether the effect size of single nucleotide polymorphisms (SNPs) on outcomes was mediated by the expression of immune-associated genes. Results The primary univariable analysis revealed a significant positive correlation between systemic lupus erythematosus (SLE), type 1 diabetes (T1D), type 2 diabetes (T2D), and rheumatoid arthritis (RA) with the risk of PE or PE with FGR. Surprisingly, a counterintuitive finding showed a significant negative association between endometriosis of pelvic peritoneum (EMoP) and the risk of PE with FGR. None of the inflammatory factors had a causal relationship with PE or PE with FGR. However, there was a significant association between lymphocyte count and the risk of PE with FGR. Within the lymphocyte subset, both the proportion of Natural Killer (NK) cells and absolute counts of naïve CD4+ T cells demonstrated significant effects on the risk of PE with FGR. Two-step MR analysis underscored the genetically predicted lymphocyte count as a significant mediator between T1D and PE with FGR. Additionally, SMR analysis indicated the potential involvement of SH2B3 in the occurrence of PE with FGR. Conclusions Our findings provided substantial evidence of the underlying causal relationship between immune dysregulation and PE or PE with FGR and some of these diseases proved to accelerate immune cells disorders and then contribute to the risk of incident PE or PE with FGR.
Collapse
Affiliation(s)
- Chumei Zeng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Huiying Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Zilian Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Jingting Li
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| |
Collapse
|
49
|
Schiavone MJ, Pérez MP, Aquieri A, Nosetto D, Pronotti MV, Mazzei M, Kudrle C, Avaca H. The Role of Obesity in the Development of Preeclampsia. Curr Hypertens Rep 2024; 26:247-258. [PMID: 38512586 DOI: 10.1007/s11906-024-01299-z] [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] [Accepted: 02/26/2024] [Indexed: 03/23/2024]
Abstract
PURPOSE OF REVIEW This comprehensive review provides an in-depth exploration of the complex relationship between obesity and preeclampsia (PE) and emphasizes the clinical implications of this association. It highlights the crucial role of screening tools in assessing individual risk and determining the need for additional antenatal care among women with obesity. The review investigates various markers for identifying the risk of developing PE, while emphasizing the significance of interventions such as exercise, weight management, and a balanced diet in reducing the incidence of preeclampsia and improving outcomes for both mother and fetus. RECENT FINDINGS Actually, there is a global pandemic of obesity, particularly among women of childbearing age and pregnant women. PE, which is characterized by maternal hypertension, proteinuria, and complications, affects 2-4% of pregnancies worldwide, posing significant risks to maternal and perinatal health. Women with obesity face an elevated risk of developing PE due to the systemic inflammation resulting from excess adiposity, which can adversely affect placental development. Adipose tissue, rich in proinflammatory cytokines and complement proteins, contributes to the pathogenesis of PE by promoting the expression of antiangiogenic factors in the mother. This review emphasizes the need for appropriate screening, interventions, and a holistic approach to reduce the incidence of preeclampsia and enhance maternal-fetal well-being, thus providing valuable insights into the multifaceted association between obesity and PE.
Collapse
Affiliation(s)
- Miguel Javier Schiavone
- Department of Hypertension, Hospital Británico de Buenos Aires, Solís 2184 - C 1134 ADT - CABA, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Mariana Paula Pérez
- Department of Hypertension, Hospital de Agudos J. M. Ramos Mejía, Ciudad Autónoma de Buenos Aires, Argentina
| | - Analía Aquieri
- Department of Hypertension, Hospital de Clínicas José de San Martín, Ciudad Autónoma de Buenos Aires, Argentina
| | - Daniela Nosetto
- Department of Hypertension, Hospital Británico de Buenos Aires, Solís 2184 - C 1134 ADT - CABA, Ciudad Autónoma de Buenos Aires, Argentina
| | - María Victoria Pronotti
- Department of Hypertension, Hospital Británico de Buenos Aires, Solís 2184 - C 1134 ADT - CABA, Ciudad Autónoma de Buenos Aires, Argentina
| | - María Mazzei
- Department of Hypertension, Hospital Británico de Buenos Aires, Solís 2184 - C 1134 ADT - CABA, Ciudad Autónoma de Buenos Aires, Argentina
| | - Cyntia Kudrle
- Department of Hypertension, Hospital Británico de Buenos Aires, Solís 2184 - C 1134 ADT - CABA, Ciudad Autónoma de Buenos Aires, Argentina
| | - Horacio Avaca
- Department of Hypertension, Hospital Británico de Buenos Aires, Solís 2184 - C 1134 ADT - CABA, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
50
|
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
|