1
|
Mack JA, Sovio U, Day FR, Gaccioli F, Cook E, Bayzid N, Cotic M, Dunton N, Madhan G, Motsinger-Reif A, Perry JRB, Charnock-Jones DS, Smith GC. Genetic Variants Associated With Preeclampsia and Maternal Serum sFLT1 Levels. Hypertension 2025; 82:839-848. [PMID: 39723542 PMCID: PMC7617282 DOI: 10.1161/hypertensionaha.124.23400] [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: 05/22/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Elevated maternal serum sFLT1 (soluble fms-like tyrosine kinase 1) has a key role in the pathophysiology of preeclampsia. We sought to determine the relationship between the maternal and fetal genome and maternal levels of sFLT1 at 12, 20, 28, and 36 weeks of gestational age (wkGA). METHODS We studied a prospective cohort of nulliparous women (3968 mother-child pairs). We related maternal and fetal genotype to the adjusted sFLT1 Z score and sFLT1:placental growth factor (PlGF) ratio Z score at each wkGA and the change in the Z score between 28 and 36 wkGA (Δ36-28). We studied genetic variants from a previous fetal genome-wide association study of preeclampsia and an externally defined polygenic score from a maternal genome-wide association study of preeclampsia. RESULTS Four variants from the fetal preeclampsia genome-wide association study were positively associated with sFLT1 and sFLT1:PlGF Z score at 36 wkGA, and FLT1 enhancer single-nucleotide polymorphisms were associated with increased Δ36-28 of sFLT1. The associations were specific for the fetal genome or stronger for the fetal than the maternal genome. An increased risk of preeclampsia based on the maternal polygenic score for preeclampsia was associated with lower levels of sFLT1 and sFLT1:PlGF ratio in the first trimester and a greater Δ36-28 for sFLT1. CONCLUSIONS The current data are consistent with a causal association between sFLT1 released by the placenta in late pregnancy and the pathophysiology of preeclampsia. The data are also consistent with maternal components to the protective effect of high sFLT1 in the first trimester and the rise in third-trimester sFLT1 levels and preeclampsia.
Collapse
Affiliation(s)
- Jasmine A. Mack
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Ulla Sovio
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- The Loke Centre for Trophoblast Research, Department of Physiology, Development, and Neuroscience, University of Cambridge, UK
| | - Felix R. Day
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Francesca Gaccioli
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- The Loke Centre for Trophoblast Research, Department of Physiology, Development, and Neuroscience, University of Cambridge, UK
| | - Emma Cook
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Nadua Bayzid
- UCL Genomics, Department of Genetics & Genomic Medicine, University College London, London, UK
| | - Marius Cotic
- UCL Genomics, Department of Genetics & Genomic Medicine, University College London, London, UK
| | - Nathan Dunton
- UCL Genomics, Department of Genetics & Genomic Medicine, University College London, London, UK
| | - Gaganjit Madhan
- UCL Genomics, Department of Genetics & Genomic Medicine, University College London, London, UK
| | - Alison Motsinger-Reif
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - John R. B. Perry
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - D. Stephen Charnock-Jones
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- The Loke Centre for Trophoblast Research, Department of Physiology, Development, and Neuroscience, University of Cambridge, UK
| | - Gordon C.S. Smith
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- The Loke Centre for Trophoblast Research, Department of Physiology, Development, and Neuroscience, University of Cambridge, UK
| |
Collapse
|
2
|
Geng J, Zhang C. Liensinine attenuates inflammatory response and oxidative stress by activation of Nrf2/HO- 1 signaling in L-NAME-induced gestational hypertension. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04105-y. [PMID: 40266297 DOI: 10.1007/s00210-025-04105-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 03/25/2025] [Indexed: 04/24/2025]
Abstract
Pregnancy-induced hypertension (PIH) is a serious condition that can affect pregnant women after 20 weeks of gestation, which leads to preeclampsia, eclampsia, or even organ damage. Liensinine exhibits a diverse array of biological activities, encompassing anti-arrhythmic, anti-cancer, anti-hypertensive, and anti-pulmonary fibrotic properties. However, the role of Liensinine remains uncharacterized in PIH. Wistar rats were induced by L-NAME to establish the PIH model. The mean arterial pressure and urine protein were measured for hypertension evaluation. The reverse transcription quantitative PCR and Western blot analyses were used for mRNA and protein levels determination, respectively. In addition, H&E staining and TUNEL staining were performed to assess histological changes. Liensinine improved hypertensive symptoms of L-NAME-induced rats by decreasing mean arterial pressure. Besides, Liensinine ameliorated the pregnancy outcomes of L-NAME-induced rats. In addition, Liensinine suppressed inflammatory response and oxidative stress injury in PIH rats. Moreover, Liensinine promoted the release of growth factors including PIGF and VEGF in PIH rats. Furthermore, Liensinine improved pathological changes of placenta tissue and inhibited cell apoptosis. Mechanistically, Liensinine activates Nrf2/HO- 1 signaling by upregulating Nrf2 and HO- 1 protein levels. Liensinine attenuated inflammatory response and oxidative stress by activation of Nrf2/HO- 1 signaling in PIH.
Collapse
Affiliation(s)
- Jie Geng
- Department of Obstetrics, Dongying People's Hospital (Dongying Hospital of Shandong Provincial Hospital Group), No.317, Nanyi Road, Dongying City, 257100, Shandong Province, China.
| | - Chunyun Zhang
- Department of Obstetrics, Dongying People's Hospital (Dongying Hospital of Shandong Provincial Hospital Group), No.317, Nanyi Road, Dongying City, 257100, Shandong Province, China
| |
Collapse
|
3
|
Ohseto H, Ishikuro M, Obara T, Narita A, Takahashi I, Shinoda G, Noda A, Murakami K, Orui M, Iwama N, Kikuya M, Metoki H, Sugawara J, Tamiya G, Kuriyama S. Preeclampsia prediction with maternal and paternal polygenic risk scores: the TMM BirThree Cohort Study. Sci Rep 2025; 15:13743. [PMID: 40258933 PMCID: PMC12012198 DOI: 10.1038/s41598-025-97291-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 04/03/2025] [Indexed: 04/23/2025] Open
Abstract
Genomic information from pregnant women and the paternal parent of their fetuses may provide effective biomarkers for preeclampsia (PE). This study investigated the association of parental polygenic risk scores (PRSs) for blood pressure (BP) and PE with PE onset and evaluated predictive performances of PRSs using clinical predictive variables. In the Tohoku Medical Megabank Project Birth and Three-Generation Cohort Study, 19,836 participants were genotyped using either Affymetrix Axiom Japonica Array v2 (further divided into two cohorts-the PRS training cohort and the internal-validation cohort-at a ratio of 1:2) or Japonica Array NEO (external-validation cohort). PRSs were calculated for systolic BP (SBP), diastolic BP (DBP), and PE and hyperparameters for PRS calculation were optimized in the training cohort. PE onset was associated with maternal SBP-, DBP-, and PE-PRSs and paternal SBP- and DBP-PRSs only in the external-validation cohort. Meta-analysis revealed overall associations with maternal PRSs but highlighted significant heterogeneity between cohorts. Maternal DBP-PRS calculated using "LDpred2" presented the most improvement in prediction models and provided additional predictive information on clinical predictive variables. Paternal DBP-PRS improved prediction models in the internal-validation cohort. In conclusion, Parental PRS, along with clinical predictive variables, is potentially useful for predicting PE.
Collapse
Affiliation(s)
- Hisashi Ohseto
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
| | - Mami Ishikuro
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan.
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan.
| | - Taku Obara
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Tohoku University Hospital, Tohoku University, Sendai, Miyagi, Japan
| | - Akira Narita
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Ippei Takahashi
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
| | - Genki Shinoda
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Aoi Noda
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Keiko Murakami
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Masatsugu Orui
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Noriyuki Iwama
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Tohoku University Hospital, Tohoku University, Sendai, Miyagi, Japan
| | - Masahiro Kikuya
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Graduate School of Medicine, Teikyo University, Itabashi-ku, Tokyo, Japan
| | - Hirohito Metoki
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Graduate School of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Junichi Sugawara
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Suzuki Memorial Hospital, Iwanuma, Miyagi, Japan
| | - Gen Tamiya
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- RIKEN Center for Advanced Intelligence Project, Chuo-ku, Tokyo, Japan
| | - Shinichi Kuriyama
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- International Research Institute of Disaster Science, Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
4
|
Mathew V, Khan RR, Jowell AR, Yan Q, Pe'er I, Truong B, Natarajan P, Yee LM, Khan SS, Sharma G, Patel AP, Cho SMJ, Pabon MA, McNeil RB, Spencer J, Silver RM, Levine LD, Grobman WA, Catov JM, Haas DM, Honigberg MC. Genetic Risk and First-Trimester Cardiovascular Health Predict Hypertensive Disorders of Pregnancy in Nulliparous Women. J Am Coll Cardiol 2025; 85:1488-1500. [PMID: 40204378 DOI: 10.1016/j.jacc.2025.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Hypertensive disorders of pregnancy (HDPs) (preeclampsia/eclampsia and gestational hypertension) are a leading cause of maternal and perinatal morbidity and mortality and are associated with long-term maternal cardiovascular disease. High genetic risk and poor cardiovascular health (CVH) are each associated with HDPs, but whether genetic risk for HDP is modified by CVH status in early pregnancy is unknown. OBJECTIVES In this study, the authors sought to test the independent and joint associations of genetic risk and first-trimester CVH with development of HDP. METHODS We examined genotyped participants from the nuMoM2b (Nulliparous Pregnancy Outcomes Study: Monitoring Mothers-to-Be), a prospective observational cohort that enrolled nulliparous individuals with singleton pregnancies from 2010 to 2013 at 8 U.S. clinical sites. Genetic risk was calculated according to a validated genetic risk score for HDP. A first-trimester CVH score was closely adapted from the American Heart Association Life's Essential 8 model. Genetic risk and CVH were each categorized as low (bottom quintile), intermediate (quintile 2-4), or high (top quintile). The primary outcome was development of HDP. Multivariable-adjusted logistic regression was used to test the independent and joint associations of genetic risk and CVH with development of HDPs. RESULTS Among 7,499 participants (mean age 27.0 years), the median first-trimester CVH score was 77.1 (Q1-Q3: 67.1-85.7). Overall, 1,032 participants (13.8%) developed an HDP (487 [6.5%] preeclampsia, 545 [7.3%] gestational hypertension). Genetic risk and CVH were each independently and additively associated with HDP (high vs low genetic risk: adjusted OR [aOR]: 2.21 [95% CI: 1.78-2.77; P < 0.001]; low vs high CVH: aOR: 2.92 [95% CI: 2.28-3.74; P < 0.001]). There was no significant interaction between genetic risk and CVH regarding risk of HDPs (Pinteraction > 0.05). HDP incidence ranged from 4.5% (low genetic risk, high CVH) to 25.7% (high genetic risk, low CVH). Compared with low CVH, high CVH was associated with 53%-74% lower risk of HDP across genetic risk strata. Findings were consistent when examining preeclampsia/eclampsia and gestational hypertension separately. CONCLUSIONS Lower genetic risk and higher first-trimester CVH were independently and additively associated with lower risk of developing HDPs in nulliparous individuals. Favorable CVH in early pregnancy may mitigate high genetic risk for HDP.
Collapse
Affiliation(s)
- Vineetha Mathew
- Tufts University School of Medicine, Boston, Massachusetts, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
| | - Raiyan R Khan
- Department of Computer Science, Columbia University, New York, New York, USA
| | - Amanda R Jowell
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Qi Yan
- Department of Computer Science, Columbia University, New York, New York, USA
| | - Itsik Pe'er
- Department of Computer Science, Columbia University, New York, New York, USA
| | - Buu Truong
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
| | - Pradeep Natarajan
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA; Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lynn M Yee
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Sadiya S Khan
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Garima Sharma
- Inova Heart and Vascular Institute, Falls Church, Virginia, USA
| | - Aniruddh P Patel
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA; Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - So Mi Jemma Cho
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
| | - Maria A Pabon
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | - Jillyn Spencer
- Intermountain Health Women and Newborn Research Department, Intermountain Healthcare, Salt Lake City, Utah, USA
| | - Robert M Silver
- Department of Obstetrics and Gynecology, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
| | - Lisa D Levine
- Pregnancy and Perinatal Research Center, Department of Obstetrics & Gynecology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - William A Grobman
- Department of Obstetrics and Gynecology, Brown University School of Medicine, Providence, Rhode Island, USA
| | - Janet M Catov
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine and Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David M Haas
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michael C Honigberg
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA; Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
5
|
Shan Y, Hu H, Chu Y. Cross-ancestry genome-wide association study identifies new susceptibility genes for preeclampsia. BMC Pregnancy Childbirth 2025; 25:379. [PMID: 40170147 PMCID: PMC11959822 DOI: 10.1186/s12884-025-07534-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Preeclampsia (PE) is a heterogeneous, multi-organ pregnancy disorder that poses a significant health burden globally, with its pathogenesis remaining unclear. This study aimed to identify novel susceptibility genes for PE through a cross-ancestry genome-wide association study (GWAS). METHODS We performed meta-analysis to summarize the PE GWAS data from the United Kingdom, Finland, and Japan. Subsequently, the multi-ancestry sum of the single-effects model was used to perform cross-ancestry fine-mapping. The functional mapping and annotation (FUMA)-expression quantitative trait loci (eQTL) mapping method, transcriptome-wide association study (TWAS)- functional summary-based imputation (FUSION) method, genome-wide complex trait analysis (GCTA)-multivariate set-based association test (mBAT)-combo method, and polygenic priority score (PoPS) method were employed to screen for candidate genes. We utilized biomarker expression level imputation using summary-level statistics (BLISS), based on summary-level protein quantitative trait loci (pQTL) data, to conduct a multi-ancestry proteome-wide association study (PWAS) analysis, followed by candidate drug prediction. RESULTS Six novel susceptibility genes associated with PE risk were identified: NPPA, SWAP70, NPR3, FGF5, REPIN1, and ACAA1. High expression of the NPPA and SWAP70 and low expression of the remaining genes were associated with a reduced risk of PE. Furthermore, we identified drugs that target NPPA, NPR3, and REPIN1. CONCLUSIONS Our study identified NPPA, SWAP70, NPR3, FGF5, REPIN1, and ACAA1 as novel genes whose predicted expression was linked to the risk of PE, offering new insights into the genetic framework of this condition.
Collapse
Affiliation(s)
- Yuping Shan
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong Hu
- Clinical Medicine, Nantong University, Nantong, China
| | - Yijing Chu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
6
|
Barragán-Zúñiga LJ, Sosa-Macias M, Simental-Mendía LE, Barragán-Zúñiga J, Lazalde-Ramos BP, Beltrán-Ontiveros S, Galaviz-Hernandez C. Association of (TG)n(GA)m repeats downstream CMA1 gene with preeclampsia in Mexican population. Placenta 2025:S0143-4004(25)00089-X. [PMID: 40199686 DOI: 10.1016/j.placenta.2025.03.018] [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: 11/29/2024] [Revised: 02/05/2025] [Accepted: 03/23/2025] [Indexed: 04/10/2025]
Abstract
Preeclampsia is a leading cause of maternal and fetal complications, often associated with endothelial dysfunction. Chymase, a proteolytic enzyme encoded by the CMA1 gene, has emerged as a potential contributor to this dysfunction. Although most preeclampsia (PE) studies have focused on maternal genetic factors, the role of paternal genetics remains underexamined. This study aimed to evaluate the association between the -1903 G/A SNV (rs1800875) and (TG)n(GA)m repeats downstream of the CMA1 gene with preeclampsia in pregnant women and their partners. A cross-sectional study was conducted involving women with PE, healthy pregnant women (HPW), and their corresponding partners, with genotyping, gene expression, and circulating protein levels assessed. A total of 141 participants were included, divided into preeclampsia (n = 80) and HPW (n = 61) groups. Women with PE showed significantly lower gestational age and higher recurrence of preeclampsia history compared to HPW. No significant association was found between the rs1800875 variant and preeclampsia; however, the (TG)n(GA)m repeat downstream of CMA1 gene was significantly associated with PE in women. Additionally, elevated serum IgE levels were significantly associated with preeclampsia (OR = 0.990; 95 % CI:0.983-0.998, p = 0.01). These findings suggest a possible role of polymorphic repeats in CMA1 as susceptibility factors for preeclampsia, indicating that both maternal and paternal genetic variations may contribute to the risk of this condition.
Collapse
Affiliation(s)
- L J Barragán-Zúñiga
- Biomedical Research Unit, Mexican Social Security Institute, Durango, Mexico
| | - M Sosa-Macias
- Academia de Genómica, Instituto Politécnico Nacional-CIIDIR, Durango, Mexico; Red Iberoamericana de Alteraciones Vasculares en Transtornos del Embarazo (RIVATREM), Chile
| | - L E Simental-Mendía
- Biomedical Research Unit, Mexican Social Security Institute, Durango, Mexico
| | - J Barragán-Zúñiga
- Programa de Doctorado en Ciencias en Biotecnología, Instituto Politécnico Nacional, Mexico
| | - B P Lazalde-Ramos
- Unidad Académica de Ciencias Químicas, Universidad Autónoma de Zacatecas, Mexico
| | - S Beltrán-Ontiveros
- Centro de Investigación y Docencia en Ciencias de la Salud (CIDOCS) de la Universidad Autónoma de Sinaloa, Mexico
| | - C Galaviz-Hernandez
- Academia de Genómica, Instituto Politécnico Nacional-CIIDIR, Durango, Mexico; Red Iberoamericana de Alteraciones Vasculares en Transtornos del Embarazo (RIVATREM), Chile.
| |
Collapse
|
7
|
Pujol Gualdo N, Džigurski J, Rukins V, Pajuste FD, Wolford BN, Võsa M, Golob M, Haug L, Alver M, Läll K, Peters M, Brumpton BM, Palta P, Mägi R, Laisk T. Atlas of genetic and phenotypic associations across 42 female reproductive health diagnoses. Nat Med 2025:10.1038/s41591-025-03543-8. [PMID: 40069456 DOI: 10.1038/s41591-025-03543-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 01/28/2025] [Indexed: 04/02/2025]
Abstract
The genetic background of many female reproductive health diagnoses remains uncharacterized, compromising our understanding of the underlying biology. Here, we map the genetic architecture across 42 female-specific health conditions using data from up to 293,618 women from two large population-based cohorts, the Estonian Biobank and the FinnGen study. Our study illustrates the utility of genetic analyses in understanding women's health better. As specific examples, we describe genetic risk factors for ovarian cysts that elucidate the genetic determinants of folliculogenesis and, by leveraging population-specific variants, uncover new candidate genes for uterine fibroids. We find that most female reproductive health diagnoses have a heritable component, with varying degrees of polygenicity and discoverability. Finally, we identify pleiotropic loci and genes that function in genital tract development (WNT4, PAX8, WT1, SALL1), hormonal regulation (FSHB, GREB1, BMPR1B, SYNE1/ESR1) and folliculogenesis (CHEK2), underlining their integral roles in female reproductive health.
Collapse
Affiliation(s)
- Natàlia Pujol Gualdo
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jelisaveta Džigurski
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Valentina Rukins
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Fanny-Dhelia Pajuste
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Brooke N Wolford
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mariann Võsa
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Mia Golob
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Lisette Haug
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Maris Alver
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kristi Läll
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Maire Peters
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Celvia CC AS, Tartu, Estonia
| | - Ben M Brumpton
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Priit Palta
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Triin Laisk
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia.
| |
Collapse
|
8
|
Lan Q, Liu S, He Q, Li Y. Changes of maternal carotid artery elasticity in patients with pregnancy concurred with hypertension. Arch Gynecol Obstet 2025; 311:731-740. [PMID: 39907747 PMCID: PMC11920326 DOI: 10.1007/s00404-025-07941-8] [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: 07/12/2024] [Accepted: 01/07/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Echo-tracking (ET) technology can visually determine whether vascular endothelial function is impaired. However, it remains uncertain whether routine ET testing for pregnant women could effectively diagnose pregnancy concurrent with hypertension (PCH) based on changes in vascular elasticity. METHODS An ET ultrasound system was employed to assess the elasticity of the carotid artery. The plasma nitric oxide (NO) levels were determined using radioimmunoassay, and the levels of endothelin-1 (ET-1) were determined using an enzyme-linked immunosorbent assay. The prediction of PCH occurrence was assessed using receiver-operating characteristic curve analysis. The correlation between the two factors was evaluated using regression analysis. RESULTS Pregnant women with PCH display significant differences in systolic blood pressure, diastolic blood pressure, and proteinuria compared to those with normal pregnancies. ET parameters, such as pulse wave velocity β (PWVβ), β-stiffness, pressure-strain elastic modulus (Eρ), and arterial compliance (AC), also reveal significant variances between women with PCH and those with normal pregnancies. These four factors are strongly associated with the disease progression in women with PCH and serve as reliable predictors for the occurrence of PCH. Additionally, the ET-1 level increases, while the NO level decreases in women with PCH, having the ability to predict the occurrence of PCH. Carotid endothelial elasticity decreases, and carotid endothelial function is impaired in pregnant women with PCH. CONCLUSION The ET parameters and indexes of carotid endothelial function can reliably predict the occurrence of PCH.
Collapse
Affiliation(s)
- Qianyu Lan
- Department of General Internal Medicine, The Fourth Hospital of Shijiazhuang, No. 16 Tangu North Street, Shijiazhuang, 050031, Hebei, China
| | - Shasha Liu
- Department of General Internal Medicine, The Fourth Hospital of Shijiazhuang, No. 16 Tangu North Street, Shijiazhuang, 050031, Hebei, China
| | - Qianqian He
- The Fifth Department of Obstetrics and Gynecology, The Fourth Hospital of Shijiazhuang, No.16 Tangu North Street, Shijiazhuang, 050031, Hebei, China.
| | - Yuan Li
- Department of General Internal Medicine, The Fourth Hospital of Shijiazhuang, No. 16 Tangu North Street, Shijiazhuang, 050031, Hebei, China.
| |
Collapse
|
9
|
Liu J, Yang Y, Wu H, Dang F, Yu X, Wang F, Wang Y, Zhao Y, Shi X, Qin W, Zhang Y, Li Y, Wang C, Shao X, Wang Y. Hypoxia-Induced O-GlcNAcylation of GATA3 Leads to Excessive Testosterone Production in Preeclamptic Placentas. MedComm (Beijing) 2025; 6:e70115. [PMID: 39991625 PMCID: PMC11847629 DOI: 10.1002/mco2.70115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 01/07/2025] [Accepted: 01/15/2025] [Indexed: 02/25/2025] Open
Abstract
The maintenance of endocrine homeostasis in the placenta is crucial for ensuring successful pregnancy. An abnormally elevated production of placental testosterone (T0) has been documented in patients with early-onset preeclamptic (E-PE). However, the underlying mechanisms remain unclear. In this study, we found that E-PE placentas exhibited significantly increased expressions of 3β-HSD1 (3β-Hydroxysteroid Dehydrogenase 1) and 17β-HSD3 (17β-Hydroxysteroid Dehydrogenase 3), the rate-limiting enzymes for T0 synthesis. This was strongly correlated with an elevated level of O-linked N-acetylglucosaminylation (O-GlcNAcylation) of GATA3 (GATA binding protein 3). In human trophoblast cells, O-linked-N-acetylglucosamine (O-GlcNAc) modification of GATA3 on Thr322 stabilized the protein and enhanced the transcriptional regulation of 3β-HSD1 and 17β-HSD3, thereby increasing T0 production. Hypoxia, a well-established pathological factor in PE, significantly enhanced the O-GlcNAcylation of GATA3 in human trophoblast cells. Our findings suggest that hypoxia-induced overactive O-GlcNAcylation of GATA3 contributes to the exacerbated T0 production in E-PE placentas. These findings provide a new perspective on the pathogenesis of E-PE from the standpoint of posttranslational regulation and may illuminate novel therapeutic strategies for adverse pregnancy outcomes such as E-PE.
Collapse
Affiliation(s)
- Juan Liu
- State Key Laboratory of Stem Cell and Reproductive BiologyKey Laboratory of Organ Regeneration and ReconstructionInstitute of ZoologyChinese Academy of SciencesBeijingChina
- Beijing Center for Disease Prevention and ControlBeijing Key Laboratory of Diagnostic and Traceability Technologies for Food PoisoningBeijingChina
| | - Yun Yang
- State Key Laboratory of Stem Cell and Reproductive BiologyKey Laboratory of Organ Regeneration and ReconstructionInstitute of ZoologyChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Hongyu Wu
- State Key Laboratory of Stem Cell and Reproductive BiologyKey Laboratory of Organ Regeneration and ReconstructionInstitute of ZoologyChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Feihong Dang
- State Key Laboratory of Stem Cell and Reproductive BiologyKey Laboratory of Organ Regeneration and ReconstructionInstitute of ZoologyChinese Academy of SciencesBeijingChina
| | - Xin Yu
- State Key Laboratory of Stem Cell and Reproductive BiologyKey Laboratory of Organ Regeneration and ReconstructionInstitute of ZoologyChinese Academy of SciencesBeijingChina
| | - Feiyang Wang
- State Key Laboratory of Stem Cell and Reproductive BiologyKey Laboratory of Organ Regeneration and ReconstructionInstitute of ZoologyChinese Academy of SciencesBeijingChina
| | - Yongqing Wang
- Department of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
| | - Yangyu Zhao
- Department of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
| | - Xiaoming Shi
- Department of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
| | - Wei Qin
- Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijingChina
| | - Yanling Zhang
- Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijingChina
| | - Yu‐Xia Li
- State Key Laboratory of Stem Cell and Reproductive BiologyKey Laboratory of Organ Regeneration and ReconstructionInstitute of ZoologyChinese Academy of SciencesBeijingChina
| | - Chu Wang
- Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijingChina
- College of Chemistry and Molecular EngineeringPeking UniversityBeijingChina
| | - Xuan Shao
- State Key Laboratory of Stem Cell and Reproductive BiologyKey Laboratory of Organ Regeneration and ReconstructionInstitute of ZoologyChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute of Stem Cell and Regenerative MedicineBeijingChina
| | - Yan‐Ling Wang
- State Key Laboratory of Stem Cell and Reproductive BiologyKey Laboratory of Organ Regeneration and ReconstructionInstitute of ZoologyChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute of Stem Cell and Regenerative MedicineBeijingChina
| |
Collapse
|
10
|
Shajihan A, Honigberg MC, Saad A, Scott NS, Powell-Wiley TM, Sharma G. Is it Time for Precision Screening in Preeclampsia? JACC. ADVANCES 2025; 4:101585. [PMID: 39938183 PMCID: PMC11870246 DOI: 10.1016/j.jacadv.2025.101585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/13/2024] [Indexed: 02/14/2025]
Affiliation(s)
- Ain Shajihan
- Inova Schar Heart and Vascular Institute, Inova Health System, Falls Church, Virginia, USA.
| | - Michael C Honigberg
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Antonio Saad
- Department of Maternal Fetal Medicine, Inova Health System, Inova Women's and Children's Hospital, Falls Church, Virginia, USA
| | - Nandita S Scott
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Tiffany M Powell-Wiley
- Cardiovascular Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA; Intramural Research Program, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, Maryland, USA
| | - Garima Sharma
- Inova Schar Heart and Vascular Institute, Inova Health System, Falls Church, Virginia, USA
| |
Collapse
|
11
|
Yang F, Zha Z, Gao F, Wang M, Du E, Wang Z, Zhou L, Gao B, Li S, Zhang D. Elucidating shared genetic association between female body mass index and preeclampsia. Commun Biol 2025; 8:322. [PMID: 40011749 DOI: 10.1038/s42003-025-07726-4] [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: 08/21/2024] [Accepted: 02/12/2025] [Indexed: 02/28/2025] Open
Abstract
The prevalence of obesity is steadily rising and poses a significant challenge to women's health. Preeclampsia (PE), a leading cause of maternal and fetal mortality, is significantly linked to a high body mass index (BMI). However, the shared genetic architecture underlying these conditions remains poorly understood. In this study, we used summary-level data from large-scale genome-wide association studies of BMI (N = 434,794) and PE (Ncases = 8185; Ncontrols = 234,147) to assess the shared genetic architecture between them. Our findings revealed a significant genetic correlation between BMI and PE, with an estimated sample overlap of approximately 0.8%. We identified roughly 1100 shared genetic variants, with the most notable region of local genetic correlation located in 16q12.2. Enrichment analyses highlighted endothelial dysfunction as a key biological mechanism linking BMI and PE. Additionally, RABEP2 was identified as a novel shared risk gene. Mendelian randomization analysis demonstrated a bidirectional causal relationship between BMI and PE, with blood pressure identified as a key mediator. We identified the shared genetic foundation between BMI and PE, providing valuable insights into the comorbidity of these conditions and offering a new framework for future research into comorbidity.
Collapse
Affiliation(s)
- Fengmei Yang
- Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Zhijian Zha
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Fang Gao
- Xiangzhou District People's Hospital, Xiangyang, China
| | - Man Wang
- Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Enfu Du
- Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Ziyang Wang
- Institute of Medicine Nursing, Hubei University of Medicine, Shiyan, China
| | - Lei Zhou
- Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Bo Gao
- Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Si Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danfeng Zhang
- Taihe Hospital, Hubei University of Medicine, Shiyan, China.
| |
Collapse
|
12
|
Martin SS, Aday AW, Allen NB, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Bansal N, Beaton AZ, Commodore-Mensah Y, Currie ME, Elkind MSV, Fan W, Generoso G, Gibbs BB, Heard DG, Hiremath S, Johansen MC, Kazi DS, Ko D, Leppert MH, Magnani JW, Michos ED, Mussolino ME, Parikh NI, Perman SM, Rezk-Hanna M, Roth GA, Shah NS, Springer MV, St-Onge MP, Thacker EL, Urbut SM, Van Spall HGC, Voeks JH, Whelton SP, Wong ND, Wong SS, Yaffe K, Palaniappan LP. 2025 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2025; 151:e41-e660. [PMID: 39866113 DOI: 10.1161/cir.0000000000001303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
BACKGROUND The American Heart Association (AHA), in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, nutrition, sleep, and obesity) and health factors (cholesterol, blood pressure, glucose control, and metabolic syndrome) that contribute to cardiovascular health. The AHA Heart Disease and Stroke Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, brain health, complications of pregnancy, kidney disease, congenital heart disease, rhythm disorders, sudden cardiac arrest, subclinical atherosclerosis, coronary heart disease, cardiomyopathy, heart failure, valvular disease, venous thromboembolism, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The AHA, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States and globally to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2025 AHA Statistical Update is the product of a full year's worth of effort in 2024 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. This year's edition includes a continued focus on health equity across several key domains and enhanced global data that reflect improved methods and incorporation of ≈3000 new data sources since last year's Statistical Update. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
13
|
Countouris M, Mahmoud Z, Cohen JB, Crousillat D, Hameed AB, Harrington CM, Hauspurg A, Honigberg MC, Lewey J, Lindley K, McLaughlin MM, Sachdev N, Sarma A, Shapero K, Sinkey R, Tita A, Wong KE, Yang E, Cho L, Bello NA. Hypertension in Pregnancy and Postpartum: Current Standards and Opportunities to Improve Care. Circulation 2025; 151:490-507. [PMID: 39960983 PMCID: PMC11973590 DOI: 10.1161/circulationaha.124.073302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Hypertension in pregnancy contributes substantially to maternal morbidity and mortality, persistent hypertension, and rehospitalization. Hypertensive disorders of pregnancy are also associated with a heightened risk of cardiovascular disease, and timely recognition and modification of associated risk factors is crucial in optimizing long-term maternal health. During pregnancy, there are expected physiologic alterations in blood pressure (BP); however, pathophysiologic alterations may also occur, leading to preeclampsia and gestational hypertension. The diagnosis and effective management of hypertension during pregnancy is essential to mitigate maternal risks, such as acute kidney injury, stroke, and heart failure, while balancing potential fetal risks, such as growth restriction and preterm birth due to altered uteroplacental perfusion. In the postpartum period, innovative and multidisciplinary care solutions that include postpartum maternal health clinics can help optimize short- and long-term care through enhanced BP management, screening of cardiovascular risk factors, and discussion of lifestyle modifications for cardiovascular disease prevention. As an adjunct to or distinct from postpartum clinics, home BP monitoring programs have been shown to improve BP ascertainment across diverse populations and to lower BP in the months after delivery. Because of concerns about pregnant patients being a vulnerable population for research, there is little evidence from trials examining the diagnosis and treatment of hypertension in pregnant and postpartum individuals. As a result, national and international guidelines differ in their recommendations, and more studies are needed to bolster future guidelines and establish best practices to achieve optimal cardiovascular health during and after pregnancy. Future research should focus on refining treatment thresholds and optimal BP range peripartum and postpartum and evaluating interventions to improve postpartum and long-term maternal cardiovascular outcomes that would advance evidence-based care and improve outcomes worldwide for people with hypertensive disorders of pregnancy.
Collapse
Affiliation(s)
| | - Zainab Mahmoud
- Department of Medicine, Division of Cardiology, Washington University in St Louis, MO
| | - Jordana B. Cohen
- Renal-Electrolyte and Hypertension Division, Department of Medicine
- Department of Biostatistics, Epidemiology, and Informatics, and Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Daniela Crousillat
- Department of Medicineand Obstetrics and Gynecology, Division of Cardiovascular Sciences, University of South Florida Morsani College of Medicine, Tampa General Hospital Heart and Vascular Institute
| | - Afshan B. Hameed
- Department of Obstetrics and Gynecology and Medicine, Division of Maternal Fetal Medicine & Cardiology, University of California, Irvine
| | - Colleen M. Harrington
- Department of Medicine, Division of Cardiology, Women’s Heart Health Program, Massachusetts General Hospital, Boston
| | - Alisse Hauspurg
- University of Pittsburgh, PA
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Alpert Medical School of Brown University, Providence, RI
| | - Michael C. Honigberg
- Department of Medicine, Division of Cardiology, Women’s Heart Health Program, Massachusetts General Hospital, Boston
| | | | - Kathryn Lindley
- Department of Medicine, Division of Cardiology, Vanderbilt University Medical Center, Nashville, TN
| | - Megan M. McLaughlin
- Department of Medicine, Division of Cardiology, University of California San Francisco
| | | | - Amy Sarma
- Department of Medicine, Division of Cardiology, Women’s Heart Health Program, Massachusetts General Hospital, Boston
| | - Kayle Shapero
- Brown University Health Cardiovascular Institute, Alpert Medical School of Brown University, Providence, RI
| | - Rachel Sinkey
- Center for Women’s Reproductive Health
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham
| | - Alan Tita
- Center for Women’s Reproductive Health
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham
| | - Kristen E. Wong
- Department of Medicine, Division of Cardiology, Washington University in St Louis, MO
| | - Eugene Yang
- Department of Medicine, Division of Cardiology, University of Washington School of Medicine, Seattle
| | - Leslie Cho
- Department of Cardiovascular Medicine, Heart Vascular Thoracic Institute at the Cleveland Clinic, OH
| | - Natalie A. Bello
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Atria Institute, New York, NY
| |
Collapse
|
14
|
Birnie K, Howe LD, Jones T, Madley-Dowd P, Martin FZ, Forbes H, Redaniel MT, Cornish R, Magnus MC, Davies NM, Tilling K, Hughes AD, Lawlor DA, Fraser A. Life course trajectories of maternal cardiovascular disease risk factors by obstetric history: a UK cohort study using electronic health records. BMC Med 2025; 23:91. [PMID: 39948598 PMCID: PMC11827161 DOI: 10.1186/s12916-025-03937-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/07/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Women who experience adverse pregnancy outcomes (APOs; gestational hypertension, preeclampsia (PE), gestational diabetes (GD), preterm birth (PTB), small or large for gestational age, miscarriage, multiple miscarriages, stillbirth, and offspring with major congenital anomalies) have increased risk of developing cardiovascular disease (CVD). We aimed to compare cardiometabolic health trajectories across the life course between women with and without APOs. METHODS We studied 187,186 women with a registered pregnancy in the UK Clinical Practice Research Datalink (CPRD) GOLD linked to Hospital Episode Statistics. Fractional polynomial multilevel models were used to compare trajectories of cardiometabolic risk factors (body mass index [BMI], blood pressure [BP], cholesterol, and glucose) between women with and without a history of APOs (individual APOs in any pregnancy and number of APOs). We explored two underlying time axes: (1) time relative to first pregnancy (from 10 years before first pregnancy to 15 years after) and (2) age. Models controlled for age at first pregnancy, residential area deprivation, non-singleton pregnancy, parity, smoking status, ethnicity, and medications use. RESULTS Women with a history of PE, gestational hypertension, or GD had higher BMI, BP, and glucose 10 years before first pregnancy compared to women without these APOs. These differences persisted 15 years post-first pregnancy. Women with a history of GD had a steeper post-partum rise in glucose. Women who experienced multiple (3 +) miscarriage, stillbirth, and/or medically indicated PTB had higher BP and BMI before and after pregnancy, with BP trajectories converging 15 years after first pregnancy. Women who experienced multiple APOs had the most adverse measurements across all cardiometabolic risk factors, with more unfavourable mean levels with each additional APO. There was little difference in cardiometabolic trajectories between women with and without a history of 1 or 2 miscarriages or congenital anomalies. CONCLUSIONS Women with APOs had adverse cardiometabolic profiles before first pregnancy, persisting up to 15 years post-pregnancy. Findings highlight the potential for targeted public health interventions to promote good cardiometabolic health in young adults transitioning from contraceptive use to planning pregnancies. APOs may identify young women who could benefit from monitoring CVD risk factors and interventions to improve cardiometabolic health.
Collapse
Affiliation(s)
- Kate Birnie
- MRC Integrative Epidemiology Unitat the , University of Bristol, Bristol, UK.
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK.
| | - Laura D Howe
- MRC Integrative Epidemiology Unitat the , University of Bristol, Bristol, UK
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Timothy Jones
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
- The National Institute for Health Research and Applied Research Collaboration West (NIHR ARC West), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
- Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, UK
| | - Paul Madley-Dowd
- MRC Integrative Epidemiology Unitat the , University of Bristol, Bristol, UK
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, Bristol, UK
| | - Florence Z Martin
- MRC Integrative Epidemiology Unitat the , University of Bristol, Bristol, UK
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Harriet Forbes
- MRC Integrative Epidemiology Unitat the , University of Bristol, Bristol, UK
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
- Faculty of Epidemiology and Population HealthandDepartment of Non-Communicable Disease EpidemiologySchool of Hygiene and Tropical Medicine, London, UK
| | - Maria Theresa Redaniel
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
- The National Institute for Health Research and Applied Research Collaboration West (NIHR ARC West), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
- National Cancer Registry Ireland, Cork, Ireland
| | - Rosie Cornish
- MRC Integrative Epidemiology Unitat the , University of Bristol, Bristol, UK
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Maria C Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Neil M Davies
- Division of Psychiatry, University College London, London, UK
- Department of Statistical Sciences, University College London, London, UK
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kate Tilling
- MRC Integrative Epidemiology Unitat the , University of Bristol, Bristol, UK
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Alun D Hughes
- MRC Unit for Lifelong Health and Ageing at University College London, London, UK
- Department of Population Science and Experimental Medicine, Institute of Cardiovascular Science, University College London, London, UK
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unitat the , University of Bristol, Bristol, UK
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Abigail Fraser
- MRC Integrative Epidemiology Unitat the , University of Bristol, Bristol, UK
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
15
|
Liu S, Guan Y, Lin S, Wu P, Zhang Q, Chu T, Dong R. Risk of Cervical Carcinoma After Unfavorable Behavior and High Genetic Risk in the UK Biobank: A Prospective Nested Case-Control Study. Biomedicines 2025; 13:464. [PMID: 40002877 PMCID: PMC11853234 DOI: 10.3390/biomedicines13020464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Previous studies have established a general understanding of the association between risky sexual behavior, genetic risk, and cervical carcinoma. However, these studies were conducted several years ago and lack systematic analysis using high-quality and population-based data. Methods: We conducted a prospective nested case-control study to identify risky behaviors and developed a behavior score. Combining the behavior score and genetic risk, we evaluated the effect of sexual and reproductive behavior and PRS on cervical carcinoma through the developed conditional logistic regression models. Results: We verified increased carcinoma risk in individuals with early sexual intercourse (OR: 1.41 [95% CI 1.09 to 1.83], p = 0.0083), non-monogamous sexual partners (OR: 3.13 [95% CI 2.15 to 4.57], p < 0.0001), three or more live births (OR: 1.44 [95% CI 1.12 to 1.84], p = 0.0040), and high PRS (polygenic risk score) (top 25% of PRS, OR: 1.58 [95% CI 1.15 to 2.16], p = 0.0044). The unfavorable sexual and reproductive behavior score we developed was linked to a 151% increased risk (OR: 2.51 [95% CI 1.79 to 3.52], p < 0.0001) after adjusting for PRS. Women with both unfavorable behavior and high genetic risk had a 5.5-fold increased cervical carcinoma risk (OR: 5.45 [95% CI 2.72 to 10.95], p < 0.0001) compared to individuals with favorable behavior and low genetic risk. Conclusions: Unfavorable sexual and reproductive behavior increases the risk of cervical carcinoma, especially in those with a high genetic risk. These findings encourage us to adhere to a healthy sexual and reproductive pattern.
Collapse
Affiliation(s)
- Shiyi Liu
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; (S.L.); (S.L.); (P.W.)
- National Clinical Research Center for Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Yunlong Guan
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China;
| | - Shitong Lin
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; (S.L.); (S.L.); (P.W.)
| | - Peng Wu
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; (S.L.); (S.L.); (P.W.)
| | - Qing Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250000, China;
- Gynecology Oncology Key Laboratory, Qilu Hospital of Shandong University, Jinan 250000, China
| | - Tian Chu
- National Clinical Research Center for Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Ruifen Dong
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250000, China;
- Gynecology Oncology Key Laboratory, Qilu Hospital of Shandong University, Jinan 250000, China
| |
Collapse
|
16
|
Jung SH, Kim H, Jung YM, Shivakumar M, Xiao B, Kim J, Jang B, Yun JS, Won HH, Park CW, Park JS, Jun JK, Kim D, Lee SM. Healthy lifestyle reduces cardiovascular risk in women with genetic predisposition to hypertensive disorders of pregnancy. Nat Commun 2025; 16:1463. [PMID: 39920105 PMCID: PMC11806095 DOI: 10.1038/s41467-025-56107-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 01/07/2025] [Indexed: 02/09/2025] Open
Abstract
The genetic risk for hypertensive disorders of pregnancy is linked with the development of atherosclerotic cardiovascular disease. However, the effects of lifestyle and metabolic syndrome on atherosclerotic cardiovascular disease have not been evaluated. Here, we assess the long-term association between these factors and atherosclerotic cardiovascular disease in women with genetic risk for hypertensive disorders of pregnancy. We evaluate the genetic risk for hypertensive disorders of pregnancy using a genome-wide polygenic risk score derived from a large-scale GWAS. The incidence of atherosclerotic cardiovascular disease is evaluated according to genetic risk, lifestyle, and metabolic syndrome. Individuals with a very high genetic risk for hypertensive disorders of pregnancy have a 53.0% higher chance of developing atherosclerotic cardiovascular disease than those with a low genetic risk. However, the risk of developing atherosclerotic cardiovascular disease is reduced by up to 64.6% through the maintenance of an ideal metabolic syndrome status and a healthy lifestyle in the high genetic risk group (top 20%), and by up to 65.4% in the low genetic risk group (bottom 20%). These findings emphasize that maintaining a healthy lifestyle in women is equally effective at reducing the risk of atherosclerotic cardiovascular disease independent of genetic risk for hypertensive disorders of pregnancy.
Collapse
Affiliation(s)
- Sang-Hyuk Jung
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Haemin Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Obstetrics and Gynecology, Kyungpook National University Chilgok Hospital, Kyungpook National University, School of Medicine, Daegu, Republic of Korea
| | - Young Mi Jung
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Manu Shivakumar
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Brenda Xiao
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jaeyoung Kim
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea
| | - Beomjin Jang
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea
| | - Jae-Seung Yun
- Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hong-Hee Won
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea
| | - Chan-Wook Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Joong Shin Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jong Kwan Jun
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dokyoon Kim
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, USA.
| | - Seung Mi Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Department of Obstetrics and Gynecology & Innovative Medical Technology Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.
- Medical Big Data Research Center & Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
17
|
Winther M, Dziegiel MH, Thorsen SU. Preeclampsia and fetal growth restriction: does novel proteomics reveal immunological possible candidate biomarkers? Curr Opin Lipidol 2025; 36:21-26. [PMID: 39607830 DOI: 10.1097/mol.0000000000000965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
PURPOSE OF REVIEW The aim of this review is to explore a possible link between immunological candidate proteins, identified through modern proteomic techniques, and preeclampsia (PE) and fetal growth restriction (FGR). RECENT FINDINGS Proteomics has become a promising tool in the search for disease pathways, drug targets, and biomarkers. PE and FGR are adverse pregnancy complications with supposed immunological involvement in their pathogenesis, but no circulating immunological biomarkers are currently established for diagnosis and risk stratification. Several proteomic studies have aimed to identify PE and FGR biomarkers - often with varying results across studies. However, proteomics has revealed altered expression of human leukocyte antigen-I in PE cases, which is supported in Genome-wide association study (GWAS) studies. Proteomic results support the heterogeneous nature of PE by identification of molecular subgroups - including subgroups characterized by immune-related proteins e.g. CXCL10. No specific immunological markers are found on FGR, but differences in overall plasma proteomic signature have been suggested. SUMMARY Proteomics certainly holds great potential. The immunological component in PE and FGR are still unclarified, but improvements in proteomic technologies may provide both definition of disease subgroups and subsequent discovery of biomarkers and targeted analysis within each subgroup.
Collapse
Affiliation(s)
- Marie Winther
- Department of Clinical Immunology, the Danish National University Hospital
| | - Morten Hanefeld Dziegiel
- Department of Clinical Immunology, the Danish National University Hospital
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
18
|
Lokki AI, Triebwasser M, Daly E, Kurki MI, Perola M, Auro K, Salmon JE, Anuja J, Daly M, Atkinson JP, Laivuori H, Meri S. Understanding rare genetic variants within the terminal pathway of complement system in preeclampsia. Genes Immun 2025; 26:22-26. [PMID: 39690307 PMCID: PMC11832413 DOI: 10.1038/s41435-024-00310-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 10/11/2024] [Accepted: 11/22/2024] [Indexed: 12/19/2024]
Abstract
Preeclampsia is a common multifactorial disease of pregnancy. Dysregulation of complement activation is among emerging candidates responsible for disease pathogenesis. In a targeted exomic sequencing study of 609 women with preeclampsia and 2092 non-preeclamptic controls, we identified 14 variants within nine genes coding for components of the membrane attack complex (MAC, C5b-9) that are associated with preeclampsia. We found two rare missense variants in the C5 gene that predispose to preeclampsia (rs200674959: I1296V, OR (CI95) = 24.13 (1.25-467.43), p value = 0.01 and rs147430470: I330T, OR (CI95) = 22.75 (1.17-440.78), p value = 0.01). In addition, one predisposing rare variant and one protective rare variant were discovered in C6 (rs41271067: D396G, OR (CI95) = 2.93 (1.18-7.10), p value = 0.01 and rs114609505: T190I, 0.02 OR (CI95) = 0.47 (0.22-0.92), p value = 0.02). The results suggest that variants in the terminal complement pathway predispose to preeclampsia.
Collapse
Affiliation(s)
- A Inkeri Lokki
- Translational Immunology Research Program, Research Programs Unit and Bacteriology and Immunology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Heart and Lung Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland.
| | - Michael Triebwasser
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Emma Daly
- Hospital and Harvard Medical School, Boston, MA, USA
| | - Mitja I Kurki
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Markus Perola
- Department of Public Health and Welfare, National Institute for Health and Welfare, Helsinki, Finland
| | - Kirsi Auro
- Department of Public Health and Welfare, National Institute for Health and Welfare, Helsinki, Finland
| | - Jane E Salmon
- Hospital for Special Surgery; Weill Medical College of Cornell University, New York, NY, USA
| | - Java Anuja
- Division of Nephrology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Mark Daly
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - John P Atkinson
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Hannele Laivuori
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital and the Wellbeing Services County of Pirkanmaa, Tampere, Finland
- Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Seppo Meri
- Translational Immunology Research Program, Research Programs Unit and Bacteriology and Immunology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- HUSLAB Diagnostic Center, Helsinki University Central Hospital, Helsinki, Finland.
| |
Collapse
|
19
|
Jiang X, Schreiner PJ, Gunderson EP, Yaffe K. Hypertensive Disorders of Pregnancy and Brain Health in Midlife: The CARDIA Study. Hypertension 2025; 82:197-205. [PMID: 39162043 PMCID: PMC11735310 DOI: 10.1161/hypertensionaha.124.22857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/30/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND To understand the role of hypertensive disorders of pregnancy (HDP), including preeclampsia and gestational hypertension (GH), in brain health earlier in life, we investigated the association of HDP with midlife cognition and brain health. METHODS We studied a prospective cohort of women, baseline age 18 to 30 years, who were assessed at study years 25 and 30 with a cognitive battery and a subset with brain magnetic resonance imaging. A history of HDP was defined based on self-report. We conducted linear regression to assess the association of a history of preeclampsia, GH, or no HDP with cognition and brain magnetic resonance imaging white matter hyperintensities. RESULTS Among 1441 women (mean age, 55.2±3.6 years), 202 reported preeclampsia and 112 reported GH. GH was associated with worse cognitive performance: global cognition (mean score, 23.2 versus 24.0; P=0.018), processing speed (67.5 versus 71.3; P=0.01), verbal fluency (29.5 versus 31.1; P=0.033), and a trend for executive function (24.3 versus 22.6; P=0.09), after multivariable adjustment. GH was associated with a greater 5-year decline in processing speed (mean change, -4.9 versus -2.7; P=0.049) and executive function (-1.7 versus 0.3; P=0.047); preeclampsia was associated with a greater 5-year decline on delayed verbal memory (-0.3 versus 0.1; P=0.041). GH and preeclampsia were associated with greater white matter hyperintensities in the parietal and frontal lobes, respectively. CONCLUSIONS GH and preeclampsia are associated with cognition and white matter hyperintensities during midlife, with differences in cognitive domains and brain lobes. Women with HDP may need to be closely monitored for adverse brain outcomes starting in midlife.
Collapse
Affiliation(s)
- Xiaqing Jiang
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco
| | | | - Erica P. Gunderson
- Division of Research, Kaiser Permanente Northern California
- Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine
| | - Kristine Yaffe
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco
- Department of Neurology, Epidemiology and Biostatistics, University of California San Francisco
- San Francisco VA Health Care System
| |
Collapse
|
20
|
Pabon MA, Weisbrod RM, Castro C, Li H, Xia P, Kang J, Ardissino M, Economy KE, Yang Z, Shi Y, Kim E, Perillo A, Barrett L, Brown JM, Divakaran S, Cetinbas M, Sadreyev RI, de Marvao A, Wood MJ, Scott NS, Lau ES, Ho JE, Di Carli MF, Roh JD, Hamburg NM, Honigberg MC. Venous Endothelial Cell Transcriptomic Profiling Implicates METAP1 in Preeclampsia. Circ Res 2025; 136:180-190. [PMID: 39727051 PMCID: PMC11747776 DOI: 10.1161/circresaha.124.324606] [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: 03/15/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Preeclampsia is a hypertensive disorder of pregnancy characterized by systemic endothelial dysfunction. The pathophysiology of preeclampsia remains incompletely understood. This study used human venous endothelial cell (EC) transcriptional profiling to investigate potential novel mechanisms underlying EC dysfunction in preeclampsia. METHODS Venous ECs were isolated from postpartum patients with severe preeclampsia and those with normotensive pregnancy using a J wire-based technique in the antecubital vein followed by CD144 (vascular endothelial cadherin) magnetic bead isolation. Venous EC transcriptomes were compared between preeclamptic and normotensive individuals. Differentially expressed genes were carried forward for genetic validation using expression quantitative trait loci from the Genotype-Tissue Expression project as exposures for vascular-specific Mendelian randomization. Functional validation of the top candidate was performed in human umbilical vein ECs using gain- and loss-of-function genetic approaches. RESULTS Seventeen individuals with preeclampsia and 7 normotensive controls were included. Pairwise analysis yielded 14 protein-coding genes nominally differentially expressed in participants with preeclampsia. Mendelian randomization revealed a significant association between higher genetically predicted METAP1 (methionyl aminopeptidase 1) expression in aortic and tibial arterial tissues and greater risk of preeclampsia. METAP1 overexpression in human umbilical vein ECs decreased angiogenesis, with a 66% decrease in tube formation (P=7.9×10-3) and 72% decrease in cell proliferation (P=2.9×10-2). Furthermore, METAP1 overexpression decreased VEGFA expression and increased expression of multiple preeclampsia-related genes, for example, FLT1, INHBA, and IL1B. Conversely, METAP1 knockdown produced opposite effects on tube formation, cell proliferation, and inflammation-related gene expression. CONCLUSIONS In a cohort of early postpartum individuals, we observed greater METAP1 expression in venous ECs of women with preeclampsia versus normotensive delivery. Mendelian randomization supported a causal relationship between greater vascular METAP1 expression and higher preeclampsia risk, and functional experiments demonstrated antiangiogenic and proinflammatory effects of METAP1 in human ECs consistent with alterations observed in preeclampsia. Ex vivo EC transcriptomics can identify novel mechanisms underlying preeclampsia pathophysiology, with implications for prevention and treatment.
Collapse
Affiliation(s)
- Maria A. Pabon
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Robert M. Weisbrod
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA
| | - Claire Castro
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
| | - Haobo Li
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA
| | - Peng Xia
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
| | - Jiayi Kang
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA
| | - Maddalena Ardissino
- British Heart Foundation, Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Philip Dahdaleh National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- National Heart and Lung Institute, Imperial College London, Hammersmith Hospital, London, UK
- Medical Research Council, Laboratory of Medical Sciences, Imperial College London, UK
| | - Katherine E. Economy
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Zihui Yang
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
| | - Yanxi Shi
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
| | | | - Anna Perillo
- Cardiovascular Imaging Program, Departments of Medicine and Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Leanne Barrett
- Cardiovascular Imaging Program, Departments of Medicine and Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Jenifer M. Brown
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Cardiovascular Imaging Program, Departments of Medicine and Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Sanjay Divakaran
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Cardiovascular Imaging Program, Departments of Medicine and Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Murat Cetinbas
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Ruslan I. Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Antonio de Marvao
- Medical Research Council, Laboratory of Medical Sciences, Imperial College London, UK
- Department of Women and Children’s Health, King’s College London, UK
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, UK
| | | | | | - Emily S. Lau
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Cardiology Division, Massachusetts General Hospital, Boston, MA
| | - Jennifer E. Ho
- CardioVascular Institute and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Marcelo F. Di Carli
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Cardiovascular Imaging Program, Departments of Medicine and Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Jason D Roh
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Cardiology Division, Massachusetts General Hospital, Boston, MA
| | - Naomi M. Hamburg
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA
| | - Michael C. Honigberg
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Cardiology Division, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
| |
Collapse
|
21
|
Kentistou KA, Lim BEM, Kaisinger LR, Steinthorsdottir V, Sharp LN, Patel KA, Tragante V, Hawkes G, Gardner EJ, Olafsdottir T, Wood AR, Zhao Y, Thorleifsson G, Day FR, Ozanne SE, Hattersley AT, O'Rahilly S, Stefansson K, Ong KK, Beaumont RN, Perry JRB, Freathy RM. Rare variant associations with birth weight identify genes involved in adipose tissue regulation, placental function and insulin-like growth factor signalling. Nat Commun 2025; 16:648. [PMID: 39809772 PMCID: PMC11733218 DOI: 10.1038/s41467-024-55761-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
Investigating the genetic factors influencing human birth weight may lead to biological insights into fetal growth and long-term health. We report analyses of rare variants that impact birth weight when carried by either fetus or mother, using whole exome sequencing data in up to 234,675 participants. Rare protein-truncating and deleterious missense variants are collapsed to perform gene burden tests. We identify 9 genes; 5 with fetal-only effects on birth weight, 1 with maternal-only effects, 3 with both, and observe directionally concordant associations in an independent sample. Four of the genes were previously implicated by GWAS of birth weight. IGF1R and PAPPA2 (fetal and maternal-acting) have known roles in insulin-like growth factor bioavailability and signalling. PPARG, INHBE and ACVR1C (fetal-acting) are involved in adipose tissue regulation, and the latter two also show associations with favourable adiposity patterns in adults. We highlight the dual role of PPARG (fetal-acting) in adipocyte differentiation and placental angiogenesis. NOS3 (fetal and maternal-acting), NRK (fetal), and ADAMTS8 (maternal-acting) have been implicated in placental function and hypertension. To conclude, our analysis of rare coding variants identifies regulators of fetal adipose tissue and fetoplacental angiogenesis as determinants of birth weight, and further evidence for the role of insulin-like growth factors.
Collapse
Affiliation(s)
- Katherine A Kentistou
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Brandon E M Lim
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Lena R Kaisinger
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | | | - Luke N Sharp
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Kashyap A Patel
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | | | - Gareth Hawkes
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Eugene J Gardner
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | | | - Andrew R Wood
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Yajie Zhao
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | | | - Felix R Day
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Susan E Ozanne
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Andrew T Hattersley
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Stephen O'Rahilly
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Kari Stefansson
- deCODE genetics/Amgen, Inc., 102 Reykjavik, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Ken K Ong
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Robin N Beaumont
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - John R B Perry
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Rachel M Freathy
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK.
| |
Collapse
|
22
|
Mack JA, Burkholder A, Akhtari FS, House JS, Sovio U, Smith GCS, Schmitt CP, Fargo DC, Hall JE, Motsinger-Reif AA. A multi-ancestry genome-wide association study identifies novel candidate loci in the RARB gene associated with hypertensive disorders of pregnancy. HGG ADVANCES 2025; 6:100385. [PMID: 39580622 PMCID: PMC11667702 DOI: 10.1016/j.xhgg.2024.100385] [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: 05/30/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
Genetic factors related to pregnancy-related traits are understudied, especially in ancestrally diverse cohorts. To assess maternal contributions to hypertensive disorders of pregnancy (HDP), we performed a multi-ancestry genome-wide association study (GWAS) of HDP in data from the North Carolina-based Personalized Environment and Genes Study (PEGS) cohort with validation in the UK Biobank (UKBB). The GWAS revealed two maternal loci associated with HDP at the genome-wide significance level. The lead independent variants were rs114954125 on chromosome 2 (near LRP1B; odds ratio [OR] [95% confidence interval {CI}]): 2.96 [2.02-4.34]; p = 2.82 × 10-8) and rs61176331 on chromosome 3 (on RARB; OR (95% CI): 3.08 (2.12-4.48); p = 3.52 × 10-9). We validated the associations near RARB with a meta-analysis of PEGS and the UKBB. We also identified cis-expression quantitative trait loci in the candidate region associated with decreased RARB expression in macrophage cells exposed to Salmonella. Chromatin mapping in FUMA identified a significant interaction within chromosome 3's enhancer and open chromatin regions, with strong effects observed for RARB and H3P10 gene regulation in mesendoderm cells, mesenchymal stem cells, and trophoblast-like stem cells. We applied existing polygenic scores (PGS) for preeclampsia and gestational hypertension and found that the scores were significantly associated with HDP in PEGS. The findings demonstrate the power of multi-ancestry studies for genetic discovery and highlight the relationship between immune response, regulation, and HDP and the utility of PGS for risk prediction. (PEGS is registered at ClinicalTrials.gov: NCT00341237.).
Collapse
Affiliation(s)
- Jasmine A Mack
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; University of Cambridge, Department of Obstetrics and Gynaecology, Cambridge CB2 0SW, UK
| | - Adam Burkholder
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Farida S Akhtari
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - John S House
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Ulla Sovio
- University of Cambridge, Department of Obstetrics and Gynaecology, Cambridge CB2 0SW, UK
| | - Gordon C S Smith
- University of Cambridge, Department of Obstetrics and Gynaecology, Cambridge CB2 0SW, UK
| | - Charles P Schmitt
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - David C Fargo
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Janet E Hall
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | | |
Collapse
|
23
|
Strauss KLE, Phoswa WN, Mokgalaboni K. The Impact of Antiretroviral Therapy on Liver Function Among Pregnant Women Living with HIV in Co-Existence with and Without Pre-Eclampsia. Viruses 2024; 17:28. [PMID: 39861817 PMCID: PMC11768528 DOI: 10.3390/v17010028] [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: 10/10/2024] [Revised: 12/09/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
Pregnant women living with HIV (PWLWHIV) are at an increased risk of developing obstetrics complications such as pre-eclampsia (PE). Antiretroviral therapy (ART) remains the standard treatment for PWLWHIV and non-pregnant women. However, its use has been associated with adverse liver conditions, particularly hepatotoxicity, often marked by elevated liver enzymes (LEEs) as demonstrated by an increased aspartate transferase (AST), alanine transaminase (ALT), and alkaline phosphatase (ALP) in PWLWHIV on ART. Morever, there is limited evidence about the effect of ART on liver function among PWLWHIV and PE. Therefore, this review examines the pathogenesis of PE and the impact of ART on liver function in PWLWHIV with and without PE. With the evidence gathered in this review, it is still unclear whether liver dysfunctions in PWLWHIV in co-existence with orwithout PE result from HIV infection or ART administration or are exacerbated by the presence of PE. Among those without PE, there was an increase in liver enzymes, a decrease, and no effect in other studies in ART-treated PWLWHIV compared to the control group. Additionally, among those with PE, the impact of ART remains unclear due to contradicting results. The notable trend was that nevirapine was associated with a reduced risk of liver dysfunction among PWLWHIV without PE. Therefore, more studies are needed in this area, especially in HIV endemic regions, to understand the exact cause of liver dysfunction in this population. This knowledge is crucial for improving liver function and PE management among PWLWHIV.
Collapse
Affiliation(s)
| | | | - Kabelo Mokgalaboni
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, Florida Campus, Roodepoort 1709, South Africa; (K.-L.E.S.); (W.N.P.)
| |
Collapse
|
24
|
Sheidu MO, Agarwala A, Lakshmanan S, Honigberg MC, Spitz JA, Sharma G. Management of pregnancy-related disorders to prevent future risk of coronary artery disease. Heart 2024; 111:83-92. [PMID: 38233160 PMCID: PMC11252244 DOI: 10.1136/heartjnl-2022-321606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2024] Open
Affiliation(s)
- Mariyam O Sheidu
- Inova Fairfax Heart and Vascular Institute, Fairfax, Virginia, USA
| | - Anandita Agarwala
- Center for Cardiovascular Disease Prevention, Baylor Scott & White The Heart Hospital Baylor Plano, Plano, Texas, USA
| | | | - Michael C Honigberg
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Garima Sharma
- Cardiology, Inova Fairfax Medical Campus, Falls Church, Virginia, USA
| |
Collapse
|
25
|
Fejzo M, Wang X, Zöllner J, Pujol-Gualdo N, Laisk T, Finer S, van Heel DA, Brumpton B, Bhatta L, Hveem K, Jasper EA, Velez Edwards DR, Hellwege JN, Edwards T, Jarvik GP, Luo Y, Khan A, MacGibbon K, Gao Y, Ge G, Averbukh I, Soon E, Angelo M, Magnus P, Johansson S, Njølstad PR, Vaudel M, Shu C, Mancuso N. Multi-ancestry GWAS of severe pregnancy nausea and vomiting identifies risk loci associated with appetite, insulin signaling, and brain plasticity. RESEARCH SQUARE 2024:rs.3.rs-5487737. [PMID: 39764105 PMCID: PMC11702859 DOI: 10.21203/rs.3.rs-5487737/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
While most pregnancies are affected by nausea and vomiting, hyperemesis gravidarum (HG) is at the severe end of the clinical spectrum and is associated with dehydration, undernutrition, and adverse maternal, fetal, and child outcomes. Herein we performed a multi-ancestry genome-wide association study (GWAS) of severe nausea and vomiting of pregnancy of 10,974 cases and 461,461 controls across European, Asian, African, and Latino ancestries. We identified ten significantly associated loci, of which six were novel (SLITRK1, SYN3, IGSF11, FSHB, TCF7L2, and CDH9), and confirmed previous genome-wide significant associations with risk genes GDF15, IGFBP7, PGR, and GFRAL. In a spatiotemporal analysis of placental development, GDF15 and TCF7L2 were expressed primarily in extra villous trophoblast, and using a weighted linear model of maternal, paternal, and fetal effects, we confirmed opposing effects for GDF15 between maternal and fetal genotype. Conversely, IGFBP7 and PGR were primarily expressed in developing maternal spiral arteries during placentation, with effects limited to the maternal genome. Risk loci were found to be under significant evolutionary selection, with the strongest effects on nausea and vomiting mid-pregnancy. Selected loci were associated with abnormal pregnancy weight gain, pregnancy duration, birth weight, head circumference, and pre-eclampsia. Potential roles for candidate genes in appetite, insulin signaling, and brain plasticity provide new pathways to explore etiological mechanisms and novel therapeutic avenues.
Collapse
Affiliation(s)
- Marlena Fejzo
- Department of Population and Public Health Science, Center for Genetic Epidemiology, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033 United States
| | - Xinran Wang
- Department of Population and Public Health Science, Center for Genetic Epidemiology, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033 United States
| | - Julia Zöllner
- UCL EGA Institute for Women's Health, University College London, London, United Kingdom
| | - Natàlia Pujol-Gualdo
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Triin Laisk
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Sarah Finer
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - David A van Heel
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - Ben Brumpton
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim 7030, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger 7600, Norway
- Clinic of Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim 7030, Norway
| | - Laxmi Bhatta
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim 7030, Norway
- Division of Mental Health Care, St Olavs Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Kristian Hveem
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim 7030, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger 7600, Norway
- Department of Research, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Elizabeth A Jasper
- Vanderbilt University Medical Center, Nashville, TN. 37221. My affiliation specifically is Department of Obstetrics and Gynecology, Division of Quantitative and Clinical Sciences
| | - Digna R Velez Edwards
- Vanderbilt University Medical Center, Nashville, TN. 37221. My affiliation specifically is Department of Obstetrics and Gynecology, Division of Quantitative and Clinical Sciences
| | - Jacklyn N Hellwege
- Vanderbilt University Medical Center, Nashville, TN. 37221. My affiliation specifically is Department of Obstetrics and Gynecology, Division of Quantitative and Clinical Sciences
| | - Todd Edwards
- Vanderbilt University Medical Center, Nashville, TN. 37221. My affiliation specifically is Department of Obstetrics and Gynecology, Division of Quantitative and Clinical Sciences
| | - Gail P Jarvik
- Departments of Medicine (Medical Genetics) and Genome Sciences, University of Washington Medical Center, Seattle, WA, USA
| | - Yuan Luo
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago IL 60611
| | - Atlas Khan
- Division of Nephrology, Dept of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY
| | - Kimber MacGibbon
- Hyperemesis Education and Research Foundation, Clackamas, OR 97089 USA
| | - Yuan Gao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031
| | - Gaoxiang Ge
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031
| | - Inna Averbukh
- Department of Pathology, Stanford University, Stanford, CA, United States
| | - Erin Soon
- Department of Pathology, Stanford University, Stanford, CA, United States
| | - Michael Angelo
- Department of Pathology, Stanford University, Stanford, CA, United States
| | - Per Magnus
- Norwegian Institute of Public Health, Oslo, Norway
| | - Stefan Johansson
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Pål R Njølstad
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Marc Vaudel
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Genetics and Bioinformatics, Health Data and Digitalization, Norwegian Institute of Public Health, Oslo, Norway
| | - Chang Shu
- Department of Population and Public Health Science, Center for Genetic Epidemiology, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033 United States
| | - Nicholas Mancuso
- Department of Population and Public Health Science, Center for Genetic Epidemiology, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033 United States
| |
Collapse
|
26
|
Honigberg MC. Classification of Myocardial Infarction in Women With Hypertensive Disorders of Pregnancy. J Am Coll Cardiol 2024; 84:2275-2277. [PMID: 39503655 DOI: 10.1016/j.jacc.2024.07.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 07/01/2024] [Indexed: 11/08/2024]
Affiliation(s)
- Michael C Honigberg
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.
| |
Collapse
|
27
|
Karlsberg RP, Cho GW, Aldana-Bitar J. A Promising Pathway Toward Mitigation and Eradication of Coronary Artery Disease. Cardiol Res 2024; 15:415-424. [PMID: 39698012 PMCID: PMC11650573 DOI: 10.14740/cr1721] [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: 08/24/2024] [Accepted: 11/08/2024] [Indexed: 12/20/2024] Open
Abstract
Cardiovascular disease remains the leading cause of death in the United States and globally. Significant advances have been made throughout the history of cardiology and the treatment of this disease; however, these efforts have not halted the alarming statistics. Emerging approaches, such as artificial intelligence applied to cardiac imaging, genetic testing, and genetic silencing, may offer essential additional steps in treating the disease. Moreover, new pathways of the disease are being identified, which differ from traditional risk factors and offer a fresh, innovative approach. This paper focuses on a novel strategy that includes identifying and treating multiple pathways of the disease using both new and traditional interventions. These interventions include plaque-directed therapy rather than surrogate therapy, with the potential to mitigate consequences and possibly eradicate the disease through personalized, multi-approach treatments similar to those used in cancer treatment.
Collapse
Affiliation(s)
- Ronald P. Karlsberg
- Cedars Sinai Heart Institute, Los Angeles, CA, USA
- University of California Los Angeles David Geffen School of Medicine, Los Angeles, CA, USA
- Cardiovascular Research Foundation of Southern California, Beverly Hills, CA, USA
| | - Geoffrey W. Cho
- University of California Los Angeles David Geffen School of Medicine, Los Angeles, CA, USA
- Cardiovascular Research Foundation of Southern California, Beverly Hills, CA, USA
| | - Jairo Aldana-Bitar
- Cardiovascular Research Foundation of Southern California, Beverly Hills, CA, USA
- The Lundquist Institute at Harbor-UCLA, Torrance, CA, USA
| |
Collapse
|
28
|
Conti-Ramsden F, de Marvao A, Gill C, Chappell LC, Myers J, Vuckovic D, Dehghan A, Hysi PG. Association of genetic ancestry with pre-eclampsia in multi-ethnic cohorts of pregnant women. Pregnancy Hypertens 2024; 38:101162. [PMID: 39368288 PMCID: PMC11870846 DOI: 10.1016/j.preghy.2024.101162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/16/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
OBJECTIVES Maternal self-reported ethnicity is recognised as a risk factor for pre-eclampsia in clinical screening tools and models. This study investigated whether ethnicity is acting as a proxy for genetic variants in this context. STUDY DESIGN A total of 436 women from multi-ethnic backgrounds recruited to two UK observational pregnancy hypertension cohort studies were genotyped. Genetically-computed individual ancestry estimates were calculated for each individual through comparison to the multi-ethnic 1000 Genomes reference panel genotypes. Regression models for pre-eclampsia using clinical risk factors including self-reported ethnicity with and without ancestry estimates were built and compared using Likelihood Ratio Tests (LRT). MAIN OUTCOME MEASURES Pre-eclampsia (early- and late-onset). RESULTS In these multi-ethnic cohorts (mean age 34.9 years; 41.3 % White, 34.2 % Black, 13.1 % Asian ethnic backgrounds; 82.6 % chronic hypertension), discrepancies between self-reported ethnicity and genetically-computed individual ancestry estimates were present in all ethnic groups, particularly minority groups. Genetically-computed pan-African ancestry percentage was associated with early-onset (< 34 weeks) pre-eclampsia in adjusted models (aOR 100 % vs 0 % African ancestry: 3.81, 95 % CI 1.04-14.14, p-value 0.044) independently of self-reported ethnicity and established clinical risk factors. Addition of genetically-computed African ancestry to a clinical risk factor model including self-reported ethnicity, improved model fit (Likelihood ratio test p-value 0.023). CONCLUSIONS Self-reported maternal ethnicity is an imperfect proxy for genetically-computed individual ancestry estimates, particularly in ethnic minority groups. Genetically-computed African ancestry percentage was associated with early-onset pre-eclampsia independently of self-reported maternal ethnicity. Well-powered studies in multi-ethnic cohorts are required to delineate the genetic contribution to pre-eclampsia.
Collapse
Affiliation(s)
- Frances Conti-Ramsden
- Department of Women and Children's Health, School of Life Course & Population Sciences, King's College London, UK.
| | - Antonio de Marvao
- Department of Women and Children's Health, School of Life Course & Population Sciences, King's College London, UK; British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, UK; Medical Research Council Laboratory of Medical Sciences, Imperial College London, UK
| | - Carolyn Gill
- Department of Women and Children's Health, School of Life Course & Population Sciences, King's College London, UK
| | - Lucy C Chappell
- Department of Women and Children's Health, School of Life Course & Population Sciences, King's College London, UK
| | - Jenny Myers
- Division of Developmental Biology and Medicine, University of Manchester, UK
| | - Dragana Vuckovic
- Department of Epidemiology and Biostatistics, Imperial College London, UK
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, Imperial College London, UK; MRC Centre for Environment and Health, School of Public Health, Imperial College London, UK; UK Dementia Research Institute, Imperial College London, UK
| | - Pirro G Hysi
- Section of Ophthalmology, School of Life Course & Population Sciences, King's College London, UK; Department of Twin Research & Genetic Epidemiology, King's College London, UK
| |
Collapse
|
29
|
Nurkkala J, Vaura F, Toivonen J, Niiranen T. Genetics of hypertension-related sex differences and hypertensive disorders of pregnancy. Blood Press 2024; 33:2408574. [PMID: 39371034 DOI: 10.1080/08037051.2024.2408574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024]
Abstract
Background: Hypertension and hypertensive disorders of pregnancy (HDP) cause a significant burden of disease on societies and individuals by increasing cardiovascular disease risk. Environmental risk factors alone do not explain the observed sexual dimorphism in lifetime blood pressure (BP) trajectories nor inter-individual variation in HDP risk. Methods: In this short review, we focus on the genetics of hypertension-related sex differences and HDP and discuss the importance of genetics utilization for sex-specific hypertension risk prediction. Results: Population and twin studies estimate that 28-66% of variation in BP levels and HDP is explained by genetic variation, while genomic wide association studies suggest that BP traits and HDP partly share a common genetic background. Moreover, environmental and epigenetic regulation of these genes differ by sex and oestrogen receptors in particular are shown to convey cardio- and vasculoprotective effects through epigenetic regulation of DNA. The majority of known genetic variation in hypertension and HDP is polygenic. Polygenic risk scores for BP display stronger associations with hypertension risk in women than in men and are associated with sex-specific age of hypertension onset. Monogenic forms of hypertension are rare and mostly present equally in both sexes. Conclusion: Despite recent genetic discoveries providing new insights into HDP and sex differences in BP traits, further research is needed to elucidate the underlying biology. Emphasis should be placed on demonstrating the added clinical value of these genetic discoveries, which may eventually facilitate genomics-based personalized treatments for hypertension and HDP.
Collapse
Affiliation(s)
- Jouko Nurkkala
- Division of Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital, Turku, Finland
- Department of Anesthesiology and Intensive Care, University of Turku, Turku, Finland
| | - Felix Vaura
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Jenni Toivonen
- Division of Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital, Turku, Finland
- Department of Anesthesiology and Intensive Care, University of Turku, Turku, Finland
| | - Teemu Niiranen
- Department of Internal Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Turku, Finland
| |
Collapse
|
30
|
Shine BK, Choi JE, Park YJ, Hong KW. The Genetic Variants Influencing Hypertension Prevalence Based on the Risk of Insulin Resistance as Assessed Using the Metabolic Score for Insulin Resistance (METS-IR). Int J Mol Sci 2024; 25:12690. [PMID: 39684400 DOI: 10.3390/ijms252312690] [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: 10/21/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Insulin resistance is a major indicator of cardiovascular diseases, including hypertension. The Metabolic Score for Insulin Resistance (METS-IR) offers a simplified and cost-effective way to evaluate insulin resistance. This study aimed to identify genetic variants associated with the prevalence of hypertension stratified by METS-IR score levels. Data from the Korean Genome and Epidemiology Study (KoGES) were analyzed. The METS-IR was calculated using the following formula: ln [(2 × fasting blood glucose (FBG) + triglycerides (TG)) × body mass index (BMI)]/ ln [high-density lipoprotein cholesterol (HDL-C)]. The participants were divided into tertiles 1 (T1) and 3 (T3) based on their METS-IR scores. Genome-wide association studies (GWAS) were performed for hypertensive cases and non-hypertensive controls within these tertile groups using logistic regression adjusted for age, sex, and lifestyle factors. Among the METS-IR tertile groups, 3517 of the 19,774 participants (17.8%) at T1 had hypertension, whereas 8653 of the 20,374 participants (42.5%) at T3 had hypertension. A total of 113 single-nucleotide polymorphisms (SNPs) reached the GWAS significance threshold (p < 5 × 10-8) in at least one tertile group, mapping to six distinct genetic loci. Notably, four loci, rs11899121 (chr2p24), rs7556898 (chr2q24.3), rs17249754 (ATP2B1), and rs1980854 (chr20p12.2), were significantly associated with hypertension in the high-METS-score group (T3). rs10857147 (FGF5) was significant in both the T1 and T3 groups, whereas rs671 (ALDH2) was significant only in the T1 group. The GWASs identified six genetic loci significantly associated with hypertension, with distinct patterns across METS-IR tertiles, highlighting the role of metabolic context in genetic susceptibility. These findings underscore critical genetic factors influencing hypertension prevalence and provide insights into the metabolic-genetic interplay underlying this condition.
Collapse
Affiliation(s)
- Bo-Kyung Shine
- Department of Family Medicine, Medical Center, Dong-A University, Busan 49201, Republic of Korea
| | - Ja-Eun Choi
- Institute of Advanced Technology, Theragen Health Co., Ltd., Seongnam 13493, Republic of Korea
| | - Young-Jin Park
- Department of Family Medicine, Medical Center, Dong-A University, Busan 49201, Republic of Korea
| | - Kyung-Won Hong
- Institute of Advanced Technology, Theragen Health Co., Ltd., Seongnam 13493, Republic of Korea
| |
Collapse
|
31
|
Fejzo M, Wang X, Zöllner J, Pujol-Gualdo N, Laisk T, Finer S, van Heel DA, Brumpton B, Bhatta L, Hveem K, Jasper EA, Velez Edwards DR, Hellwege JN, Edwards T, Jarvik GP, Luo Y, Khan A, MacGibbon K, Gao Y, Ge G, Averbukh I, Soon E, Angelo M, Magnus P, Johansson S, Njølstad PR, Vaudel M, Shu C, Mancuso N. Multi-ancestry GWAS of severe pregnancy nausea and vomiting identifies risk loci associated with appetite, insulin signaling, and brain plasticity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.19.24317559. [PMID: 39606329 PMCID: PMC11601681 DOI: 10.1101/2024.11.19.24317559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
While most pregnancies are affected by nausea and vomiting, hyperemesis gravidarum (HG) is at the severe end of the clinical spectrum and is associated with dehydration, undernutrition, and adverse maternal, fetal, and child outcomes. Herein we performed a multi-ancestry genome-wide association study (GWAS) of severe nausea and vomiting of pregnancy of 10,974 cases and 461,461 controls across European, Asian, African, and Latino ancestries. We identified ten significantly associated loci, of which six were novel (SLITRK1, SYN3, IGSF11, FSHB, TCF7L2, and CDH9), and confirmed previous genome-wide significant associations with risk genes GDF15, IGFBP7, PGR, and GFRAL. In a spatiotemporal analysis of placental development, GDF15 and TCF7L2 were expressed primarily in extra villous trophoblast, and using a weighted linear model of maternal, paternal, and fetal effects, we confirmed opposing effects for GDF15 between maternal and fetal genotype. Conversely, IGFBP7 and PGR were primarily expressed in developing maternal spiral arteries during placentation, with effects limited to the maternal genome. Risk loci were found to be under significant evolutionary selection, with the strongest effects on nausea and vomiting mid-pregnancy. Selected loci were associated with abnormal pregnancy weight gain, pregnancy duration, birth weight, head circumference, and pre-eclampsia. Potential roles for candidate genes in appetite, insulin signaling, and brain plasticity provide new pathways to explore etiological mechanisms and novel therapeutic avenues.
Collapse
Affiliation(s)
- Marlena Fejzo
- Department of Population and Public Health Science, Center for Genetic Epidemiology, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033 United States
| | - Xinran Wang
- Department of Population and Public Health Science, Center for Genetic Epidemiology, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033 United States
| | - Julia Zöllner
- UCL EGA Institute for Women's Health, University College London, London, United Kingdom
| | - Natàlia Pujol-Gualdo
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Triin Laisk
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Sarah Finer
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - David A van Heel
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - Ben Brumpton
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim 7030, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger 7600, Norway
- Clinic of Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim 7030, Norway
| | - Laxmi Bhatta
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim 7030, Norway
- Division of Mental Health Care, St Olavs Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Kristian Hveem
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim 7030, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger 7600, Norway
- Department of Research, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Elizabeth A Jasper
- Vanderbilt University Medical Center, Nashville, TN. 37221. My affiliation specifically is Department of Obstetrics and Gynecology, Division of Quantitative and Clinical Sciences
| | - Digna R Velez Edwards
- Vanderbilt University Medical Center, Nashville, TN. 37221. My affiliation specifically is Department of Obstetrics and Gynecology, Division of Quantitative and Clinical Sciences
| | - Jacklyn N Hellwege
- Vanderbilt University Medical Center, Nashville, TN. 37221. My affiliation specifically is Department of Obstetrics and Gynecology, Division of Quantitative and Clinical Sciences
| | - Todd Edwards
- Vanderbilt University Medical Center, Nashville, TN. 37221. My affiliation specifically is Department of Obstetrics and Gynecology, Division of Quantitative and Clinical Sciences
| | - Gail P Jarvik
- Departments of Medicine (Medical Genetics) and Genome Sciences, University of Washington Medical Center, Seattle, WA, USA
| | - Yuan Luo
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago IL 60611
| | - Atlas Khan
- Division of Nephrology, Dept of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY
| | - Kimber MacGibbon
- Hyperemesis Education and Research Foundation, Clackamas, OR 97089 USA
| | - Yuan Gao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031
| | - Gaoxiang Ge
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031
| | - Inna Averbukh
- Department of Pathology, Stanford University, Stanford, CA, United States
| | - Erin Soon
- Department of Pathology, Stanford University, Stanford, CA, United States
| | - Michael Angelo
- Department of Pathology, Stanford University, Stanford, CA, United States
| | - Per Magnus
- Norwegian Institute of Public Health, Oslo, Norway
| | - Stefan Johansson
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Pål R Njølstad
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Marc Vaudel
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Genetics and Bioinformatics, Health Data and Digitalization, Norwegian Institute of Public Health, Oslo, Norway
| | - Chang Shu
- Department of Population and Public Health Science, Center for Genetic Epidemiology, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033 United States
| | - Nicholas Mancuso
- Department of Population and Public Health Science, Center for Genetic Epidemiology, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033 United States
| |
Collapse
|
32
|
Carbonneau M, Li Y, Qu Y, Zheng Y, Wood AC, Wang M, Liu C, Huan T, Joehanes R, Guo X, Yao J, Taylor KD, Tracy RP, Peter D, Liu Y, Johnson WC, Post WS, Blackwell T, Rotter JI, Rich SS, Redline S, Fornage M, Wang J, Ning H, Hou L, Lloyd-jones D, Ferrier K, Min YI, Carson AP, Raffield LM, Teumer A, Grabe HJ, Völzke H, Nauck M, Dörr M, Domingo-Relloso A, Fretts A, Tellez-Plaza M, Cole S, Navas-Acien A, Wang M, Murabito JM, Heard-Costa NL, Prescott B, Xanthakis V, Mozaffarian D, Levy D, Ma J. DNA Methylation Signatures of Cardiovascular Health Provide Insights into Diseases. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.19.24317587. [PMID: 39606375 PMCID: PMC11601778 DOI: 10.1101/2024.11.19.24317587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background The association of overall cardiovascular health (CVH) with changes in DNA methylation (DNAm) has not been well characterized. Methods We calculated the American Heart Association's Life's Essential 8 (LE8) score to reflect CVH in five cohorts with diverse ancestry backgrounds. Epigenome-wide association studies (EWAS) for LE8 score were conducted, followed by bioinformatic analyses. DNAm loci significantly associated with LE8 score were used to calculate a CVH DNAm score. We examined the association of the CVH DNAm score with incident CVD, CVD-specific mortality, and all-cause mortality. Results We identified 609 CpGs associated with LE8 score at false discovery rate (FDR) < 0.05 in the discovery analysis and at Bonferroni corrected P < 0.05 in the multi-cohort replication stage. Most had low-to-moderate heterogeneity (414 CpGs [68.0%] with I2 < 0.2) in replication analysis. Pathway enrichment analyses and phenome-wide association study (PheWAS) search associated these CpGs with inflammatory or autoimmune phenotypes. We observed enrichment for phenotypes in the EWAS catalog, with 29-fold enrichment for stroke (P = 2.4e-15) and 21-fold for ischemic heart disease (P = 7.4e-38). Two-sample Mendelian randomization (MR) analysis showed significant association between 141 CpGs and ten phenotypes (261 CpG-phenotype pairs) at FDR < 0.05. For example, hypomethylation at cg20544516 (MIR33B; SREBF1) associated with lower risk of stroke (P = 8.1e-6). In multivariable prospective analyses, the CVH DNAm score was consistently associated with clinical outcomes across participating cohorts, the reduction in risk of incident CVD, CVD mortality, and all-cause mortality per standard deviation increase in the DNAm score ranged from 19% to 32%, 28% to 40%, and 27% to 45%, respectively. Conclusions We identified new DNAm signatures for CVH across diverse cohorts. Our analyses indicate that immune response-related pathways may be the key mechanism underpinning the association between CVH and clinical outcomes.
Collapse
Affiliation(s)
- Madeleine Carbonneau
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
- Framingham Heart Study, Framingham, MA
| | - Yi Li
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Yishu Qu
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 N Lake Shore Drive, Chicago, IL 60611, USA
| | - Yinan Zheng
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 N Lake Shore Drive, Chicago, IL 60611, USA
| | - Alexis C. Wood
- United States Department of Agriculture (USDA)/ARS Children’s Nutrition Research Center, Baylor College of Medicine, TX, USA
| | - Mengyao Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Chunyu Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Tianxiao Huan
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
- Framingham Heart Study, Framingham, MA
| | - Roby Joehanes
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
- Framingham Heart Study, Framingham, MA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 W. Carson Street, Torrance, CA 90502, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 W. Carson Street, Torrance, CA 90502, USA
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 W. Carson Street, Torrance, CA 90502, USA
| | - Russell P. Tracy
- Department of Pathology & Laboratory Medicine, University of Vermont Larner College of Medicine, 360 South Park Drive, Colchester, VT 05446, USA
| | - Durda Peter
- Department of Pathology & Laboratory Medicine, University of Vermont Larner College of Medicine, 360 South Park Drive, Colchester, VT 05446, USA
| | - Yongmei Liu
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - W Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Wendy S. Post
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Tom Blackwell
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 W. Carson Street, Torrance, CA 90502, USA
| | - Stephen S. Rich
- Department of Genome Sciences, University of Virginia School of Medicine, 1200 Jefferson Park Avenue, Charlottesville, VA 22903, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham & Women’s Hospital & Harvard Medical School, Boston, MA, 02115, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, TX 77030, USA
| | - Jun Wang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 N Lake Shore Drive, Chicago, IL 60611, USA
| | - Hongyan Ning
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 N Lake Shore Drive, Chicago, IL 60611, USA
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 N Lake Shore Drive, Chicago, IL 60611, USA
| | - Donald Lloyd-jones
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 N Lake Shore Drive, Chicago, IL 60611, USA
| | - Kendra Ferrier
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Aurora, CO 80045, USA
| | - Yuan-I. Min
- Department of Medicine, University of Mississippi Medical Center, 350 W. Woodrow Wilson Avenue, Suite 701, Jackson, MS 39213, USA
| | - April P. Carson
- Department of Medicine, University of Mississippi Medical Center, 350 W. Woodrow Wilson Avenue, Suite 701, Jackson, MS 39213, USA
| | - Laura M. Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Alexander Teumer
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Hans J. Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Partner Site Rostock/Greifswald, Greifswald, Germany
| | - Henry Völzke
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department SHIP/Clinical-Epidemiological Research, Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Matthias Nauck
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Marcus Dörr
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Arce Domingo-Relloso
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, USA
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Amanda Fretts
- Department of Epidemiology, Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, USA
| | - Maria Tellez-Plaza
- Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Carlos III Health Institute, Madrid, Spain
| | - Shelley Cole
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Ana Navas-Acien
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Meng Wang
- Nutrition Epidemiology and Data Science, Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
| | - Joanne M. Murabito
- Framingham Heart Study, Framingham, MA
- Department of Medicine, Section of General Internal Medicine Boston University Chobanian & Avedisian School of Medicine, Boston, MA and Boston Medical Center, Boston, MA
| | - Nancy L. Heard-Costa
- Department of Medicine, Section of General Internal Medicine Boston University Chobanian & Avedisian School of Medicine, Boston, MA and Boston Medical Center, Boston, MA
| | - Brenton Prescott
- Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, MA
| | - Vanessa Xanthakis
- Framingham Heart Study, Framingham, MA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
- Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, MA
| | - Dariush Mozaffarian
- Nutrition Epidemiology and Data Science, Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
| | - Daniel Levy
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
- Framingham Heart Study, Framingham, MA
| | - Jiantao Ma
- Nutrition Epidemiology and Data Science, Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
| |
Collapse
|
33
|
Dasinger JH, Abais-Battad JM, Walton SD, Burns-Ray EC, Cherian-Shaw M, Baldwin KE, Fehrenbach DJ, Mattson DL. Intact NOX2 in T Cells Mediates Pregnancy-Induced Renal Damage in Dahl SS Rats. Hypertension 2024; 81:2357-2367. [PMID: 39301728 PMCID: PMC11517830 DOI: 10.1161/hypertensionaha.124.23303] [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: 05/07/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Hypertensive disorders of pregnancy are associated with increased risk for cardiovascular disease, renal disease, and mortality. While the exact mechanisms remain unclear, T cells and reactive oxygen species have been implicated in its pathogenesis. We utilized Dahl salt-sensitive (SS), SSCD247-/- (Dahl SS CD247 knockout rat; lacking T cells), and SSp67phox-/- (Dahl SS p67phox [NOX2 (NADPH [nitcotinamide adenine dinucleotide phosphate] oxidase 2)] knockout rat; lacking NOX2) rats to investigate these mechanisms in primigravida and multigravida states. METHODS We assessed blood pressure and renal damage phenotypes in SS, SSCD247-/-, and SSp67phox-/- rats during primigravida and multigravida states. To investigate the contribution of NOX2 in T cells, we performed adoptive transfers of splenocytes or cluster of differentiation (CD)4+ T cells from either SS or SSp67phox-/- donors into SSCD247-/- recipients to determine pregnancy-specific alterations in phenotype. RESULTS Multigravida SS rats developed significant pregnancy-induced renal damage and renal functional impairment associated with elevated maternal mortality rates, whereas deletion of T cells or NOX2 garnered protection. During primigravida states, this attenuation in renal damage was observed, with the greatest protection in the SSp67phox-/- rat. To demonstrate that NOX2 in T cells contributes to adverse pregnancy phenotypes, adoptive transfer of SS splenocytes into SSCD247-/- rats resulted in significant pregnancy-induced renal damage, whereas transfer of SSp67phox-/- splenocytes garnered protection. Specifically, the transfer of SS CD4+ T cells resulted in pregnancy-induced proteinuria and increases in uterine artery resistance index, an effect not seen with the transfer of SSp67phox-/- CD4+ T cells. CONCLUSIONS T cells and NOX2-derived reactive oxygen species, thus, contribute to end-organ damage in both primigravida and multigravida pregnancies in the SS rat leading to increases in maternal mortality.
Collapse
Affiliation(s)
- John Henry Dasinger
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta GA
| | | | - Samuel D Walton
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta GA
| | - Emily C Burns-Ray
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta GA
| | - Mary Cherian-Shaw
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta GA
| | - Kaitlyn E Baldwin
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta GA
| | | | - David L. Mattson
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta GA
| |
Collapse
|
34
|
Capalbo A, de Wert G, Mertes H, Klausner L, Coonen E, Spinella F, Van de Velde H, Viville S, Sermon K, Vermeulen N, Lencz T, Carmi S. Screening embryos for polygenic disease risk: a review of epidemiological, clinical, and ethical considerations. Hum Reprod Update 2024; 30:529-557. [PMID: 38805697 PMCID: PMC11369226 DOI: 10.1093/humupd/dmae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/25/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND The genetic composition of embryos generated by in vitro fertilization (IVF) can be examined with preimplantation genetic testing (PGT). Until recently, PGT was limited to detecting single-gene, high-risk pathogenic variants, large structural variants, and aneuploidy. Recent advances have made genome-wide genotyping of IVF embryos feasible and affordable, raising the possibility of screening embryos for their risk of polygenic diseases such as breast cancer, hypertension, diabetes, or schizophrenia. Despite a heated debate around this new technology, called polygenic embryo screening (PES; also PGT-P), it is already available to IVF patients in some countries. Several articles have studied epidemiological, clinical, and ethical perspectives on PES; however, a comprehensive, principled review of this emerging field is missing. OBJECTIVE AND RATIONALE This review has four main goals. First, given the interdisciplinary nature of PES studies, we aim to provide a self-contained educational background about PES to reproductive specialists interested in the subject. Second, we provide a comprehensive and critical review of arguments for and against the introduction of PES, crystallizing and prioritizing the key issues. We also cover the attitudes of IVF patients, clinicians, and the public towards PES. Third, we distinguish between possible future groups of PES patients, highlighting the benefits and harms pertaining to each group. Finally, our review, which is supported by ESHRE, is intended to aid healthcare professionals and policymakers in decision-making regarding whether to introduce PES in the clinic, and if so, how, and to whom. SEARCH METHODS We searched for PubMed-indexed articles published between 1/1/2003 and 1/3/2024 using the terms 'polygenic embryo screening', 'polygenic preimplantation', and 'PGT-P'. We limited the review to primary research papers in English whose main focus was PES for medical conditions. We also included papers that did not appear in the search but were deemed relevant. OUTCOMES The main theoretical benefit of PES is a reduction in lifetime polygenic disease risk for children born after screening. The magnitude of the risk reduction has been predicted based on statistical modelling, simulations, and sibling pair analyses. Results based on all methods suggest that under the best-case scenario, large relative risk reductions are possible for one or more diseases. However, as these models abstract several practical limitations, the realized benefits may be smaller, particularly due to a limited number of embryos and unclear future accuracy of the risk estimates. PES may negatively impact patients and their future children, as well as society. The main personal harms are an unindicated IVF treatment, a possible reduction in IVF success rates, and patient confusion, incomplete counselling, and choice overload. The main possible societal harms include discarded embryos, an increasing demand for 'designer babies', overemphasis of the genetic determinants of disease, unequal access, and lower utility in people of non-European ancestries. Benefits and harms will vary across the main potential patient groups, comprising patients already requiring IVF, fertile people with a history of a severe polygenic disease, and fertile healthy people. In the United States, the attitudes of IVF patients and the public towards PES seem positive, while healthcare professionals are cautious, sceptical about clinical utility, and concerned about patient counselling. WIDER IMPLICATIONS The theoretical potential of PES to reduce risk across multiple polygenic diseases requires further research into its benefits and harms. Given the large number of practical limitations and possible harms, particularly unnecessary IVF treatments and discarded viable embryos, PES should be offered only within a research context before further clarity is achieved regarding its balance of benefits and harms. The gap in attitudes between healthcare professionals and the public needs to be narrowed by expanding public and patient education and providing resources for informative and unbiased genetic counselling.
Collapse
Affiliation(s)
- Antonio Capalbo
- Juno Genetics, Department of Reproductive Genetics, Rome, Italy
- Center for Advanced Studies and Technology (CAST), Department of Medical Genetics, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Guido de Wert
- Department of Health, Ethics & Society, CAPHRI-School for Public Health and Primary Care and GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Heidi Mertes
- Department of Philosophy and Moral Sciences, Ghent University, Ghent, Belgium
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | - Liraz Klausner
- Braun School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Edith Coonen
- Departments of Clinical Genetics and Reproductive Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
- School for Oncology and Developmental Biology, GROW, Maastricht University, Maastricht, The Netherlands
| | - Francesca Spinella
- Eurofins GENOMA Group Srl, Molecular Genetics Laboratories, Department of Scientific Communication, Rome, Italy
| | - Hilde Van de Velde
- Research Group Genetics Reproduction and Development (GRAD), Vrije Universiteit Brussel, Brussel, Belgium
- Brussels IVF, UZ Brussel, Brussel, Belgium
| | - Stephane Viville
- Laboratoire de Génétique Médicale LGM, Institut de Génétique Médicale d’Alsace IGMA, INSERM UMR 1112, Université de Strasbourg, France
- Laboratoire de Diagnostic Génétique, Unité de Génétique de l’infertilité (UF3472), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Karen Sermon
- Research Group Genetics Reproduction and Development (GRAD), Vrije Universiteit Brussel, Brussel, Belgium
| | | | - Todd Lencz
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Departments of Psychiatry and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Shai Carmi
- Braun School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
35
|
Ardissino M, Truong B, Slob EAW, Schuermans A, Yoshiji S, Morley AP, Burgess S, Ng FS, de Marvao A, Natarajan P, Nicolaides K, Gaziano L, Butterworth A, Honigberg MC. Proteome- and Transcriptome-Wide Genetic Analysis Identifies Biological Pathways and Candidate Drug Targets for Preeclampsia. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004755. [PMID: 39119725 PMCID: PMC7616531 DOI: 10.1161/circgen.124.004755] [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: 04/05/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Preeclampsia is a leading cause of maternal and perinatal morbidity and mortality. However, the current understanding of its underlying biological pathways remains limited. METHODS In this study, we performed a cross-platform proteome- and transcriptome-wide genetic analysis aimed at evaluating the causal relevance of >2000 circulating proteins with preeclampsia, supported by data on the expression of over 15 000 genes across 36 tissues leveraging large-scale preeclampsia genetic association data from women of European ancestry. RESULTS We demonstrate genetic associations of 18 circulating proteins with preeclampsia (SULT1A1 [sulfotransferase 1A1], SH2B3 [SH2B adapter protein 3], SERPINE2 [serpin family E member 2], RGS18 [regulator of G-protein signaling 18], PZP [pregnancy zone protein], NOTUM [notum, palmitoleoyl-protein carboxylesterase], METAP1 [methionyl aminopeptidase 1], MANEA [mannosidase endo-alpha], jun-D [JunD proto-oncogene], GDF15 [growth differentiation factor 15], FGL1 [fibrinogen like 1], FGF5 [fibroblast growth factor 5], FES [FES proto-oncogene], APOBR [apolipoprotein B receptor], ANP [natriuretic peptide A], ALDH-E2 [aldehyde dehydrogenase 2 family member], ADAMTS13 [ADAM metallopeptidase with thrombospondin type 1 motif 13], and 3MG [N-methylpurine DNA glycosylase]), among which 11 were either directly or indirectly supported by gene expression data, 9 were supported by Bayesian colocalization analyses, and 5 (SERPINE2, PZP, FGF5, FES, and ANP) were supported by all lines of evidence examined. Protein interaction mapping identified potential shared biological pathways through natriuretic peptide signaling, blood pressure regulation, immune tolerance, and thrombin activity regulation. CONCLUSIONS This investigation identified multiple targetable proteins linked to cardiovascular, inflammatory, and coagulation pathways, with SERPINE2, PZP, FGF5, FES, and ANP identified as pivotal proteins with likely causal roles in the development of preeclampsia. The identification of these potential targets may guide the development of targeted therapies for preeclampsia.
Collapse
Affiliation(s)
- Maddalena Ardissino
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (M.A., L.G., A.B.), University of Cambridge, United Kingdom
- Victor Phillip Dahdaleh Heart and Lung Research Institute (M.A, A.B.), University of Cambridge, United Kingdom
- National Heart and Lung Institute (M.A, F.S.N.), Imperial College London, United Kingdom
- Medical Research Council, London Institute of Medical Sciences (M.A.), Imperial College London, United Kingdom
| | - Buu Truong
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative (B.T., A.S., P.N., M.C.H.)
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (B.T., A.S., P.N., M.C.H.)
| | - Eric A W Slob
- MRC Biostatistics Unit (E.A.W.S., S.B.), University of Cambridge, United Kingdom
- Department of Applied Economics, Erasmus School of Economics (E.A.W.S.), Erasmus University Rotterdam, the Netherlands
- Erasmus University Rotterdam Institute for Behavior and Biology (E.A.W.S.), Erasmus University Rotterdam, the Netherlands
- Department of Psychology, Education and Child Studies (E.A.W.S.), Erasmus University Rotterdam, the Netherlands
| | - Art Schuermans
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative (B.T., A.S., P.N., M.C.H.)
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (B.T., A.S., P.N., M.C.H.)
- Faculty of Medicine, KU Leuven, Belgium (A.S.)
| | - Satoshi Yoshiji
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA (S.Y.)
| | - Alec P Morley
- Department of Medicine, School of Clinical Medicine (A.P.M.), University of Cambridge, United Kingdom
- Gonville and Caius College (A.P.M.), University of Cambridge, United Kingdom
| | - Stephen Burgess
- MRC Biostatistics Unit (E.A.W.S., S.B.), University of Cambridge, United Kingdom
| | - Fu Siong Ng
- National Heart and Lung Institute (M.A, F.S.N.), Imperial College London, United Kingdom
| | - Antonio de Marvao
- Department of Women and Children's Health (A.d.M., K.N.)
- British Heart Foundation Center of Research Excellence, School of Cardiovascular Medicine and Sciences (A.d.M.), King's College Hospital, London, United Kingdom
- Fetal Medicine Research Institute (A.d.M., K.N.), King's College Hospital, London, United Kingdom
| | - Pradeep Natarajan
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative (B.T., A.S., P.N., M.C.H.)
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (B.T., A.S., P.N., M.C.H.)
| | - Kypros Nicolaides
- Department of Women and Children's Health (A.d.M., K.N.)
- Fetal Medicine Research Institute (A.d.M., K.N.), King's College Hospital, London, United Kingdom
| | - Liam Gaziano
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (M.A., L.G., A.B.), University of Cambridge, United Kingdom
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System (L.G.), Harvard Medical School, Boston
| | - Adam Butterworth
- Victor Phillip Dahdaleh Heart and Lung Research Institute (M.A, A.B.), University of Cambridge, United Kingdom
- British Heart Foundation Center of Research Excellence (A.B.), University of Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus (A.B.), University of Cambridge, United Kingdom
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics (A.B.), University of Cambridge, United Kingdom
| | - Michael C Honigberg
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative (B.T., A.S., P.N., M.C.H.)
- Department of Medicine (M.C.H.), Harvard Medical School, Boston
| |
Collapse
|
36
|
Ali M, Ahmed M, Memon M, Chandio F, Shaikh Q, Parveen A, Phull AR. Preeclampsia: A comprehensive review. Clin Chim Acta 2024; 563:119922. [PMID: 39142550 DOI: 10.1016/j.cca.2024.119922] [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: 07/03/2024] [Revised: 08/11/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Preeclampsia (PE) is a life-threatening disease of pregnancy and a prominent cause of neonatal and maternal mortality and morbidity. PE affects approximately 5-10% of pregnancies worldwide, posing significant risks to perinatal and maternal health. It is characterized by a variety of interconnected pathological cascades contributing to the stimulation of intravascular inflammation, oxidative stress (OS), endothelial cell activation, and syncytiotrophoblast stress that converge on a common pathway, ultimately resulting in disease progression. The present study was designed and executed to review the existing scientific literature, specifically focusing on the etiology (gestational diabetes mellitus and maternal obesity, insulin resistance, metabolic syndrome, maternal infection, periodontal disease, altered microbiome, and genetics), clinical presentations (hypertension, blood disorders, proteinuria, hepatic dysfunction, renal dysfunction, pulmonary edema, cardiac dysfunction, fetal growth restrictions, and eclampsia), therapeutic clinical biomarkers (creatinine, albuminuria, and cystatin C) along with their associations and mechanisms in PE. In addition, this study provides insights into the potential of nanomedicines for targeting these mechanisms for PE management and treatment. Inflammation, OS, proteinuria, and an altered microbiome are prominent biomarkers associated with progression and PE-related pathogenesis. Understanding the molecular mechanisms, exploring suitable markers, targeted interventions, comprehensive screening, and holistic strategies are critical to decreasing the incidence of PE and promoting maternal-fetal well-being. The present study comprehensively reviewed the etiology, clinical presentations, therapeutic biomarkers, and preventive potential of nanomedicines in the treatment and management of PE.
Collapse
Affiliation(s)
- Majida Ali
- Department of Gynecology and Obstetrics, Shaikh Zaid Women Hospital Larkana, Shaheed Mohtarma Benazir Bhutto Medical University (SMBB) Larkana, Pakistan
| | - Madiha Ahmed
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Jaffer Khan Jamali Road, H-8/4, Islamabad, Pakistan
| | - Mehwish Memon
- Department of Biochemistry, Ibn e Sina University, Mirpur Khas, Pakistan
| | - Fozia Chandio
- Department of Gynecology and Obstetrics, Shaikh Zaid Women Hospital Larkana, Shaheed Mohtarma Benazir Bhutto Medical University (SMBB) Larkana, Pakistan
| | - Quratulain Shaikh
- Department of Gynecology and Obstetrics, Shaikh Zaid Women Hospital Larkana, Shaheed Mohtarma Benazir Bhutto Medical University (SMBB) Larkana, Pakistan
| | - Amna Parveen
- College of Pharmacy, Gachon University, No. 191, Hambakmoero, Yeonsu-gu, Incheon 21936, South Korea.
| | - Abdul-Rehman Phull
- Department of Biochemistry, Shah Abdul Latif University, Khairpur, Sindh, Pakistan.
| |
Collapse
|
37
|
Kovacheva VP, Venkatachalam S, Pfister C, Anwer T. Preeclampsia and eclampsia: Enhanced detection and treatment for morbidity reduction. Best Pract Res Clin Anaesthesiol 2024; 38:246-256. [PMID: 39764814 PMCID: PMC11707392 DOI: 10.1016/j.bpa.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/18/2024] [Accepted: 11/15/2024] [Indexed: 01/11/2025]
Abstract
Preeclampsia is a life-threatening complication that develops in 2-8% of pregnancies. It is characterized by elevated blood pressure after 20 weeks of gestation and may progress to multiorgan dysfunction, leading to severe maternal and fetal morbidity and mortality. The only definitive treatment is delivery, and efforts are focused on early risk prediction, surveillance, and severity mitigation. Anesthesiologists, as part of the interdisciplinary team, should evaluate patients early in labor in order to optimize cardiovascular, pulmonary, and coagulation status. Neuraxial techniques are safe in the absence of coagulopathy and aid avoidance of general anesthesia, which is associated with high risk in these patients. This review aims to provide anaesthesiologists with a comprehensive update on the latest strategies and evidence-based practices for managing preeclampsia, with an emphasis on perioperative care.
Collapse
Affiliation(s)
- Vesela P Kovacheva
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, L1, Boston, MA, 02115, USA.
| | - Shakthi Venkatachalam
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, L1, Boston, MA, 02115, USA.
| | - Claire Pfister
- UCT Department of Anaesthesia and Perioperative Medicine, Groote Schuur Hospital, Main Road, Observatory, Cape Town, Postal code 7935, South Africa.
| | - Tooba Anwer
- Division of Maternal-Fetal Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, L1, Boston, MA, 02115, USA.
| |
Collapse
|
38
|
Cao C, Saxena R, Gray KJ. Placental Origins of Preeclampsia: Insights from Multi-Omic Studies. Int J Mol Sci 2024; 25:9343. [PMID: 39273292 PMCID: PMC11395466 DOI: 10.3390/ijms25179343] [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: 07/22/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Preeclampsia (PE) is a major cause of maternal and neonatal morbidity and mortality worldwide, with the placenta playing a central role in disease pathophysiology. This review synthesizes recent advancements in understanding the molecular mechanisms underlying PE, focusing on placental genes, proteins, and genetic variants identified through multi-omic approaches. Transcriptomic studies in bulk placental tissue have identified many dysregulated genes in the PE placenta, including the PE signature gene, Fms-like tyrosine kinase 1 (FLT1). Emerging single-cell level transcriptomic data have revealed key cell types and molecular signatures implicated in placental dysfunction and PE. However, the considerable variability among studies underscores the need for standardized methodologies and larger sample sizes to enhance the reproducibility of results. Proteomic profiling of PE placentas has identified numerous PE-associated proteins, offering insights into potential biomarkers and pathways implicated in PE pathogenesis. Despite significant progress, challenges such as inconsistencies in study findings and lack of validation persist. Recent fetal genome-wide association studies have identified multiple genetic loci associated with PE, with ongoing efforts to elucidate their impact on placental gene expression and function. Future directions include the integration of multi-omic data, validation of findings in diverse PE populations and clinical subtypes, and the development of analytical approaches and experimental models to study the complex interplay of placental and maternal factors in PE etiology. These insights hold promise for improving risk prediction, diagnosis, and management of PE, ultimately reducing its burden on maternal and neonatal health.
Collapse
Affiliation(s)
- Chang Cao
- Center for Genomic Medicine and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Richa Saxena
- Center for Genomic Medicine and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Kathryn J. Gray
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
39
|
Fry H, Mazidi M, Kartsonaki C, Clarke R, Walters RG, Chen Z, Millwood IY. The Role of Furin and Its Therapeutic Potential in Cardiovascular Disease Risk. Int J Mol Sci 2024; 25:9237. [PMID: 39273186 PMCID: PMC11394739 DOI: 10.3390/ijms25179237] [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: 07/12/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/15/2024] Open
Abstract
Furin is an important proteolytic enzyme, converting several proteins from inactive precursors to their active forms. Recently, proteo-genomic analyses in European and East Asian populations suggested a causal association of furin with ischaemic heart disease, and there is growing interest in its role in cardiovascular disease (CVD) aetiology. In this narrative review, we present a critical appraisal of evidence from population studies to assess furin's role in CVD risk and potential as a drug target for CVD. Whilst most observational studies report positive associations between furin expression and CVD risk, some studies report opposing effects, which may reflect the complex biological roles of furin and its substrates. Genetic variation in FURIN is also associated with CVD and its risk factors. We found no evidence of current clinical development of furin as a drug target for CVD, although several phase 1 and 2 clinical trials of furin inhibitors as a type of cancer immunotherapy have been completed. The growing field of proteo-genomics in large-scale population studies may inform the future development of furin and other potential drug targets to improve the treatment and prevention of CVD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Iona Y. Millwood
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK; (H.F.); (M.M.); (C.K.); (R.C.); (R.G.W.); (Z.C.)
| |
Collapse
|
40
|
Jasper EA, Hellwege JN, Breeyear JH, Xiao B, Jarvik GP, Stanaway IB, Leppig KA, Chittoor G, Hayes MG, Dikilitas O, Kullo IJ, Holm IA, Verma SS, Edwards TL, Velez Edwards DR. Genetic predictors of blood pressure traits are associated with preeclampsia. Sci Rep 2024; 14:17613. [PMID: 39080328 PMCID: PMC11289248 DOI: 10.1038/s41598-024-68469-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 07/24/2024] [Indexed: 08/02/2024] Open
Abstract
Preeclampsia, a pregnancy complication characterized by hypertension after 20 gestational weeks, is a major cause of maternal and neonatal morbidity and mortality. Mechanisms leading to preeclampsia are unclear; however, there is evidence of high heritability. We evaluated the association of polygenic scores (PGS) for blood pressure traits and preeclampsia to assess whether there is shared genetic architecture. Non-Hispanic Black and White reproductive age females with pregnancy indications and genotypes were obtained from Vanderbilt University's BioVU, Electronic Medical Records and Genomics network, and Penn Medicine Biobank. Preeclampsia was defined by ICD codes. Summary statistics for diastolic blood pressure (DBP), systolic blood pressure (SBP), and pulse pressure (PP) PGS were acquired from Giri et al. Associations between preeclampsia and each PGS were evaluated separately by race and data source before subsequent meta-analysis. Ten-fold cross validation was used for prediction modeling. In 3504 Black and 5009 White included individuals, the rate of preeclampsia was 15.49%. In cross-ancestry meta-analysis, all PGSs were associated with preeclampsia (ORDBP = 1.10, 95% CI 1.02-1.17, p = 7.68 × 10-3; ORSBP = 1.16, 95% CI 1.09-1.23, p = 2.23 × 10-6; ORPP = 1.14, 95% CI 1.07-1.27, p = 9.86 × 10-5). Addition of PGSs to clinical prediction models did not improve predictive performance. Genetic factors contributing to blood pressure regulation in the general population also predispose to preeclampsia.
Collapse
Affiliation(s)
- Elizabeth A Jasper
- Division of Quantitative and Clinical Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, 2525 West End Avenue, Suite 600, Rm 616, Nashville, TN, 37203, USA
- Center for Precision Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Epidemiology Center, Vanderbilt University, Nashville, TN, USA
- Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jacklyn N Hellwege
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Epidemiology Center, Vanderbilt University, Nashville, TN, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joseph H Breeyear
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Brenda Xiao
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Gail P Jarvik
- Departments of Medicine (Medical Genetics) and Genome Sciences, University of Washington Medical Center, Seattle, WA, USA
| | - Ian B Stanaway
- Division of Nephrology and Harborview Medical Center Kidney Research Institute, Department of Medicine, University of Washington Medical Center, Seattle, WA, USA
| | | | - Geetha Chittoor
- Department of Population Health Sciences, Geisinger, Danville, PA, USA
| | - M Geoffrey Hayes
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Anthropology, Northwestern University, Evanston, IL, USA
| | - Ozan Dikilitas
- Departments of Internal Medicine, Cardiovascular Medicine, Mayo Clinician-Investigator Training Program, Mayo Clinic, Rochester, MN, USA
| | - Iftikhar J Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ingrid A Holm
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Shefali Setia Verma
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Todd L Edwards
- Vanderbilt Epidemiology Center, Vanderbilt University, Nashville, TN, USA
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Digna R Velez Edwards
- Division of Quantitative and Clinical Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, 2525 West End Avenue, Suite 600, Rm 616, Nashville, TN, 37203, USA.
- Vanderbilt Epidemiology Center, Vanderbilt University, Nashville, TN, USA.
- Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, USA.
| |
Collapse
|
41
|
Juybar M, Shahraki M, Ghasemi M, Payandeh A, Saljooghi S, Saravani M. A Comparison of Apelin Rs56204867 and Apelin Receptor Rs11544374 Gene Polymorphisms and Their Association with Risk of Preeclampsia in Southeast Iran. Rep Biochem Mol Biol 2024; 13:273-280. [PMID: 39995647 PMCID: PMC11847591 DOI: 10.61186/rbmb.13.2.273] [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: 07/10/2024] [Accepted: 11/24/2024] [Indexed: 02/26/2025]
Abstract
Background Pre-eclampsia (PE) is a severe pregnancy condition with genetic and environmental factors affecting the placental function and vascular changes. Genetic variants in the apelinergic system may influence preeclampsia risk and birth outcomes. Therefore, this study aimed to compare apelin (APLN) rs56204867 and apelin receptor (APLNR) rs11544374 gene polymorphisms and to investigate their association with mothers' body mass index and infant's birth weight among women with preeclampsia and control group in southeast Iran. Methods A total of 123 PE patients and 125 age- and gender-matched control subjects were enrolled in the study. The PCR-RFLP method was employed to genotype the APLN rs56204867 and APLNR rs11544374 gene polymorphisms. Results There was no significant association between the genotypes of the rs11544374 variant and the PE risk. The incidence of the AG genotype of the rs54204867 variant in the control group was considerably greater than in the PE group. Also, a significant relationship was found between the body mass profile of patients with PE and the APLN rs54204867 gene polymorphism. Conclusions It was observed that the APLN rs54204867 gene polymorphism could affect the PE risk. No significant difference was found between the PE group and the control group in terms of the genotypes of the APLNR rs11544374 variant. It was not statistically significant between mothers' BMI and rs11544374 of the APLNR gene, whereas an obvious link was observed between mothers' BMI and rs54204867 of the APLN gene.
Collapse
Affiliation(s)
- Maryam Juybar
- Department of Nutrition, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Mansour Shahraki
- Department of Nutrition, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
- Adolescent Health Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Marzieh Ghasemi
- Department of Obstetrics and Gynecology, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Abolfazl Payandeh
- Department of Biostatistics and Epidemiology, Genetics of Non-Communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Shaghayegh Saljooghi
- Genetics of Non-communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Mohsen Saravani
- Genetics of Non-communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| |
Collapse
|
42
|
Tschiderer L, van der Schouw YT, Burgess S, Bloemenkamp KWM, Seekircher L, Willeit P, Onland-Moret C, Peters SAE. Hypertensive disorders of pregnancy and cardiovascular disease risk: a Mendelian randomisation study. Heart 2024; 110:710-717. [PMID: 38148158 PMCID: PMC11103302 DOI: 10.1136/heartjnl-2023-323490] [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: 09/22/2023] [Accepted: 11/27/2023] [Indexed: 12/28/2023] Open
Abstract
OBJECTIVE Observational studies show that hypertensive disorders of pregnancy (HDPs) are related to unfavourable maternal cardiovascular disease (CVD) risk profiles later in life. We investigated whether genetic liability to pre-eclampsia/eclampsia and gestational hypertension is associated with CVD risk factors and occurrence of CVD events. METHODS We obtained genetic associations with HDPs from a genome-wide association study and used individual participant data from the UK Biobank to obtain genetic associations with CVD risk factors and CVD events (defined as myocardial infarction or stroke). In our primary analysis, we applied Mendelian randomisation using inverse-variance weighted regression analysis in ever pregnant women. In sensitivity analyses, we studied men and nulligravidae to investigate genetic liability to HDPs and CVD risk without the ability to experience the underlying phenotype. RESULTS Our primary analysis included 221 155 ever pregnant women (mean age 56.8 (SD 7.9) years) with available genetic data. ORs for CVD were 1.20 (1.02 to 1.41) and 1.24 (1.12 to 1.38) per unit increase in the log odds of genetic liability to pre-eclampsia/eclampsia and gestational hypertension, respectively. Furthermore, genetic liability to HDPs was associated with higher levels of systolic and diastolic blood pressure and younger age at hypertension diagnosis. Sensitivity analyses revealed no statistically significant differences when comparing the findings with those of nulligravidae and men. CONCLUSIONS Genetic liability to HDPs is associated with higher CVD risk, lower blood pressure levels and earlier hypertension diagnosis. Our study suggests similar findings in ever pregnant women, nulligravidae and men, implying biological mechanisms relating to HDPs are causally related to CVD risk.
Collapse
Affiliation(s)
- Lena Tschiderer
- Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Kitty W M Bloemenkamp
- Department of Obstetrics, Division Women and Baby, Birth Centre, Wilhelmina Children Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Lisa Seekircher
- Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria
| | - Peter Willeit
- Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Charlotte Onland-Moret
- Julius Center for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Sanne A E Peters
- Julius Center for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht, The Netherlands
- The George Institute for Global Health, School of Public Health, Imperial College London, London, UK
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
43
|
Kentistou KA, Lim BEM, Kaisinger LR, Steinthorsdottir V, Sharp LN, Patel KA, Tragante V, Hawkes G, Gardner EJ, Olafsdottir T, Wood AR, Zhao Y, Thorleifsson G, Day FR, Ozanne SE, Hattersley AT, O'Rahilly S, Stefansson K, Ong KK, Beaumont RN, Perry JRB, Freathy RM. Rare variant associations with birth weight identify genes involved in adipose tissue regulation, placental function and insulin-like growth factor signalling. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.03.24305248. [PMID: 38633783 PMCID: PMC11023655 DOI: 10.1101/2024.04.03.24305248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Investigating the genetic factors influencing human birth weight may lead to biological insights into fetal growth and long-term health. Genome-wide association studies of birth weight have highlighted associated variants in more than 200 regions of the genome, but the causal genes are mostly unknown. Rare genetic variants with robust evidence of association are more likely to point to causal genes, but to date, only a few rare variants are known to influence birth weight. We aimed to identify genes that harbour rare variants that impact birth weight when carried by either the fetus or the mother, by analysing whole exome sequence data in UK Biobank participants. We annotated rare (minor allele frequency <0.1%) protein-truncating or high impact missense variants on whole exome sequence data in up to 234,675 participants with data on their own birth weight (fetal variants), and up to 181,883 mothers who reported the birth weight of their first child (maternal variants). Variants within each gene were collapsed to perform gene burden tests and for each associated gene, we compared the observed fetal and maternal effects. We identified 8 genes with evidence of rare fetal variant effects on birth weight, of which 2 also showed maternal effects. One additional gene showed evidence of maternal effects only. We observed 10/11 directionally concordant associations in an independent sample of up to 45,622 individuals (sign test P=0.01). Of the genes identified, IGF1R and PAPPA2 (fetal and maternal-acting) have known roles in insulin-like growth factor bioavailability and signalling. PPARG, INHBE and ACVR1C (all fetal-acting) have known roles in adipose tissue regulation and rare variants in the latter two also showed associations with favourable adiposity patterns in adults. We highlight the dual role of PPARG in both adipocyte differentiation and placental angiogenesis. NOS3, NRK, and ADAMTS8 (fetal and maternal-acting) have been implicated in both placental function and hypertension. Analysis of rare coding variants has identified regulators of fetal adipose tissue and fetoplacental angiogenesis as determinants of birth weight, as well as further evidence for the role of insulin-like growth factors.
Collapse
Affiliation(s)
- Katherine A Kentistou
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Brandon E M Lim
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Lena R Kaisinger
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | | | - Luke N Sharp
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Kashyap A Patel
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | | | - Gareth Hawkes
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Eugene J Gardner
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | | | - Andrew R Wood
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Yajie Zhao
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | | | - Felix R Day
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Susan E Ozanne
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Andrew T Hattersley
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Stephen O'Rahilly
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Kari Stefansson
- deCODE genetics/Amgen, Inc., 102 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Ken K Ong
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Robin N Beaumont
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - John R B Perry
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Rachel M Freathy
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
44
|
Lokki A, Triebwasser M, Daly E, Cohort F, Kurki M, Perola M, Auro K, Salmon J, Java A, Daly M, Atkinson J, Laivuori H, Meri S. Rare variants in genes coding for components of the terminal pathway of the complement system in preeclampsia. RESEARCH SQUARE 2024:rs.3.rs-4121735. [PMID: 38645143 PMCID: PMC11030519 DOI: 10.21203/rs.3.rs-4121735/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Preeclampsia is a common multifactorial disease of pregnancy. Dysregulation of the complement activation is among emerging candidates responsible for disease pathogenesis. In a targeted exomic sequencing study we identified 14 variants within nine genes coding for components of the membrane attack complex (MAC, C5b-9) that are associated with preeclampsia. We found two rare missense variants in the C5 gene that predispose to preeclampsia (rs200674959: I1296V, OR (CI95) = 24.13 (1.25-467.43), p-value = 0.01 and rs147430470: I330T, OR (CI95) = 22.75 (1.17-440.78), p-value = 0.01). In addition, one predisposing rare variant and one protective rare variant were discovered in C6 (rs41271067: D396G, OR (CI95) = 2.93 (1.18-7.10), p-value = 0.01 and rs114609505: T190I, 0.02 OR (CI95) = 0.47 (0.22-0.92), p-value = 0.02). The results suggest that variants in terminal complement pathway predispose to preeclampsia.
Collapse
|
45
|
Lindley KJ, Perry A, Jacobs M, Petty L, Amancherla K, Zhao S, Barker C, Davila-Roman VG, Khan SS, Osmundson SS, Tanriverdi K, Freedman JE, Below J, Shah RV, Laurent LC. Differences in Cardiometabolic Proteins in Pregnancy Prioritize Relevant Targets of Preeclampsia. Arterioscler Thromb Vasc Biol 2024; 44:969-975. [PMID: 38385288 DOI: 10.1161/atvbaha.124.320737] [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: 01/17/2024] [Accepted: 02/05/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Preeclampsia is a hypertensive disorder of pregnancy characterized by widespread vascular inflammation. It occurs frequently in pregnancy, often without known risk factors, and has high rates of maternal and fetal morbidity and mortality. Identification of biomarkers that predict preeclampsia and its cardiovascular sequelae before clinical onset, or even before pregnancy, is a critical unmet need for the prevention of adverse pregnancy outcomes. METHODS We explored differences in cardiovascular proteomics (Olink Explore 384) in 256 diverse pregnant persons across 2 centers (26% Hispanic, 21% Black). RESULTS We identified significant differences in plasma abundance of markers associated with angiogenesis, blood pressure, cell adhesion, inflammation, and metabolism between individuals delivering with preeclampsia and controls, some of which have not been widely described previously and are not represented in the preeclampsia placental transcriptome. While we observed a broadly similar pattern in early (<34 weeks) versus late (≥34 weeks) preeclampsia, several proteins related to hemodynamic stress, hemostasis, and immune response appeared to be more highly dysregulated in early preeclampsia relative to late preeclampsia. CONCLUSIONS These results demonstrate the value of performing targeted proteomics using a panel of cardiovascular biomarkers to identify biomarkers relevant to preeclampsia pathophysiology and highlight the need for larger multiomic studies to define modifiable pathways of surveillance and intervention upstream to preeclampsia diagnosis.
Collapse
Affiliation(s)
- Kathryn J Lindley
- Vanderbilt Translational and Clinical Research Center, Cardiovascular Division (K.J.L., A.P., K.A., S.Z., K.T., J.E.F., R.V.S.), Vanderbilt University Medical Center, Nashville, TN
- Department of Obstetrics and Gynecology (K.J.L., S.S.O.), Vanderbilt University Medical Center, Nashville, TN
| | - Andrew Perry
- Vanderbilt Translational and Clinical Research Center, Cardiovascular Division (K.J.L., A.P., K.A., S.Z., K.T., J.E.F., R.V.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Marni Jacobs
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal Fetal Medicine, University of California San Diego (M.J.)
| | - Lauren Petty
- Division of Genetic Medicine (L.P., J.B.), Vanderbilt University Medical Center, Nashville, TN
| | - Kaushik Amancherla
- Vanderbilt Translational and Clinical Research Center, Cardiovascular Division (K.J.L., A.P., K.A., S.Z., K.T., J.E.F., R.V.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Shilin Zhao
- Vanderbilt Translational and Clinical Research Center, Cardiovascular Division (K.J.L., A.P., K.A., S.Z., K.T., J.E.F., R.V.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Claire Barker
- Cardiovascular Imaging and Clinical Research Core Laboratory, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (C.B., V.G.D.-R.)
| | - Victor G Davila-Roman
- Cardiovascular Imaging and Clinical Research Core Laboratory, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO (C.B., V.G.D.-R.)
| | - Sadiya S Khan
- Cardiovascular Division, Feinberg School of Medicine, Northwestern University, Chicago, IL (S.S.K.)
| | - Sarah S Osmundson
- Department of Obstetrics and Gynecology (K.J.L., S.S.O.), Vanderbilt University Medical Center, Nashville, TN
| | - Kahraman Tanriverdi
- Vanderbilt Translational and Clinical Research Center, Cardiovascular Division (K.J.L., A.P., K.A., S.Z., K.T., J.E.F., R.V.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Jane E Freedman
- Vanderbilt Translational and Clinical Research Center, Cardiovascular Division (K.J.L., A.P., K.A., S.Z., K.T., J.E.F., R.V.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Jennifer Below
- Division of Genetic Medicine (L.P., J.B.), Vanderbilt University Medical Center, Nashville, TN
| | - Ravi V Shah
- Vanderbilt Translational and Clinical Research Center, Cardiovascular Division (K.J.L., A.P., K.A., S.Z., K.T., J.E.F., R.V.S.), Vanderbilt University Medical Center, Nashville, TN
| | | |
Collapse
|
46
|
Yang ML, Xu C, Gupte T, Hoffmann TJ, Iribarren C, Zhou X, Ganesh SK. Sex-specific genetic architecture of blood pressure. Nat Med 2024; 30:818-828. [PMID: 38459180 PMCID: PMC11797078 DOI: 10.1038/s41591-024-02858-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/05/2024] [Indexed: 03/10/2024]
Abstract
The genetic and genomic basis of sex differences in blood pressure (BP) traits remain unstudied at scale. Here, we conducted sex-stratified and combined-sex genome-wide association studies of BP traits using the UK Biobank resource, identifying 1,346 previously reported and 29 new BP trait-associated loci. Among associated loci, 412 were female-specific (Pfemale ≤ 5 × 10-8; Pmale > 5 × 10-8) and 142 were male-specific (Pmale ≤ 5 × 10-8; Pfemale > 5 × 10-8); these sex-specific loci were enriched for hormone-related transcription factors, in particular, estrogen receptor 1. Analyses of gene-by-sex interactions and sexually dimorphic effects identified four genomic regions, showing female-specific associations with diastolic BP or pulse pressure, including the chromosome 13q34-COL4A1/COL4A2 locus. Notably, female-specific pulse pressure-associated loci exhibited enriched acetylated histone H3 Lys27 modifications in arterial tissues and a female-specific association with fibromuscular dysplasia, a female-biased vascular disease; colocalization signals included Chr13q34: COL4A1/COL4A2, Chr9p21: CDKN2B-AS1 and Chr4q32.1: MAP9 regions. Sex-specific and sex-biased polygenic associations of BP traits were associated with multiple cardiovascular traits. These findings suggest potentially clinically significant and BP sex-specific pleiotropic effects on cardiovascular diseases.
Collapse
Affiliation(s)
- Min-Lee Yang
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Chang Xu
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Trisha Gupte
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Thomas J Hoffmann
- Department of Epidemiology & Biostatistics, and Institute for Human Genetics, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | - Xiang Zhou
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Santhi K Ganesh
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA.
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA.
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
47
|
Schuermans A, Truong B, Ardissino M, Bhukar R, Slob EAW, Nakao T, Dron JS, Small AM, Cho SMJ, Yu Z, Hornsby W, Antoine T, Lannery K, Postupaka D, Gray KJ, Yan Q, Butterworth AS, Burgess S, Wood MJ, Scott NS, Harrington CM, Sarma AA, Lau ES, Roh JD, Januzzi JL, Natarajan P, Honigberg MC. Genetic Associations of Circulating Cardiovascular Proteins With Gestational Hypertension and Preeclampsia. JAMA Cardiol 2024; 9:209-220. [PMID: 38170504 PMCID: PMC10765315 DOI: 10.1001/jamacardio.2023.4994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 11/01/2023] [Indexed: 01/05/2024]
Abstract
Importance Hypertensive disorders of pregnancy (HDPs), including gestational hypertension and preeclampsia, are important contributors to maternal morbidity and mortality worldwide. In addition, women with HDPs face an elevated long-term risk of cardiovascular disease. Objective To identify proteins in the circulation associated with HDPs. Design, Setting, and Participants Two-sample mendelian randomization (MR) tested the associations of genetic instruments for cardiovascular disease-related proteins with gestational hypertension and preeclampsia. In downstream analyses, a systematic review of observational data was conducted to evaluate the identified proteins' dynamics across gestation in hypertensive vs normotensive pregnancies, and phenome-wide MR analyses were performed to identify potential non-HDP-related effects associated with the prioritized proteins. Genetic association data for cardiovascular disease-related proteins were obtained from the Systematic and Combined Analysis of Olink Proteins (SCALLOP) consortium. Genetic association data for the HDPs were obtained from recent European-ancestry genome-wide association study meta-analyses for gestational hypertension and preeclampsia. Study data were analyzed October 2022 to October 2023. Exposures Genetic instruments for 90 candidate proteins implicated in cardiovascular diseases, constructed using cis-protein quantitative trait loci (cis-pQTLs). Main Outcomes and Measures Gestational hypertension and preeclampsia. Results Genetic association data for cardiovascular disease-related proteins were obtained from 21 758 participants from the SCALLOP consortium. Genetic association data for the HDPs were obtained from 393 238 female individuals (8636 cases and 384 602 controls) for gestational hypertension and 606 903 female individuals (16 032 cases and 590 871 controls) for preeclampsia. Seventy-five of 90 proteins (83.3%) had at least 1 valid cis-pQTL. Of those, 10 proteins (13.3%) were significantly associated with HDPs. Four were robust to sensitivity analyses for gestational hypertension (cluster of differentiation 40, eosinophil cationic protein [ECP], galectin 3, N-terminal pro-brain natriuretic peptide [NT-proBNP]), and 2 were robust for preeclampsia (cystatin B, heat shock protein 27 [HSP27]). Consistent with the MR findings, observational data revealed that lower NT-proBNP (0.76- to 0.88-fold difference vs no HDPs) and higher HSP27 (2.40-fold difference vs no HDPs) levels during the first trimester of pregnancy were associated with increased risk of HDPs, as were higher levels of ECP (1.60-fold difference vs no HDPs). Phenome-wide MR analyses identified 37 unique non-HDP-related protein-disease associations, suggesting potential on-target effects associated with interventions lowering HDP risk through the identified proteins. Conclusions and Relevance Study findings suggest genetic associations of 4 cardiovascular disease-related proteins with gestational hypertension and 2 associated with preeclampsia. Future studies are required to test the efficacy of targeting the corresponding pathways to reduce HDP risk.
Collapse
Affiliation(s)
- Art Schuermans
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Buu Truong
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
| | - Maddalena Ardissino
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Rohan Bhukar
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
| | - Eric A. W. Slob
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
- Department of Applied Economics, Erasmus School of Economics, Erasmus University Rotterdam, Rotterdam, the Netherlands
- Erasmus University Rotterdam Institute for Behavior and Biology, Erasmus University Rotterdam, Rotterdam, the Netherlands
| | - Tetsushi Nakao
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Jacqueline S. Dron
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
| | - Aeron M. Small
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - So Mi Jemma Cho
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
- Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Zhi Yu
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
| | - Whitney Hornsby
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
| | - Tajmara Antoine
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
| | - Kim Lannery
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
| | - Darina Postupaka
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
| | - Kathryn J. Gray
- Division of Maternal-Fetal Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Qi Yan
- Department of Obstetrics and Gynecology, Columbia University, New York, New York
| | - Adam S. Butterworth
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- BHF Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, United Kingdom
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
| | - Malissa J. Wood
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiology Division, Massachusetts General Hospital, Boston
- Lee Health, Fort Myers, Florida
| | - Nandita S. Scott
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiology Division, Massachusetts General Hospital, Boston
| | - Colleen M. Harrington
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiology Division, Massachusetts General Hospital, Boston
| | - Amy A. Sarma
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiology Division, Massachusetts General Hospital, Boston
| | - Emily S. Lau
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiology Division, Massachusetts General Hospital, Boston
| | - Jason D. Roh
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiology Division, Massachusetts General Hospital, Boston
| | - James L. Januzzi
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiology Division, Massachusetts General Hospital, Boston
- Baim Institute for Clinical Research, Boston, Massachusetts
| | - Pradeep Natarajan
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiology Division, Massachusetts General Hospital, Boston
| | - Michael C. Honigberg
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiology Division, Massachusetts General Hospital, Boston
| |
Collapse
|
48
|
Hu J, Chen L, Ruan J, Chen X. The role of the annexin A protein family at the maternal-fetal interface. Front Endocrinol (Lausanne) 2024; 15:1314214. [PMID: 38495790 PMCID: PMC10940358 DOI: 10.3389/fendo.2024.1314214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 02/09/2024] [Indexed: 03/19/2024] Open
Abstract
Successful pregnancy requires the tolerance of the maternal immune system for the semi-allogeneic embryo, as well as a synchrony between the receptive endometrium and the competent embryo. The annexin family belongs to calcium-regulated phospholipid-binding protein, which functions as a membrane skeleton to stabilize the lipid bilayer and participate in various biological processes in humans. There is an abundance of the annexin family at the maternal-fetal interface, and it exerts a crucial role in embryo implantation and the subsequent development of the placenta. Altered expression of the annexin family and dysfunction of annexin proteins or polymorphisms of the ANXA gene are involved in a range of pregnancy complications. In this review, we summarize the current knowledge of the annexin A protein family at the maternal-fetal interface and its association with female reproductive disorders, suggesting the use of ANXA as the potential therapeutic target in the clinical diagnosis and treatment of pregnancy complications.
Collapse
Affiliation(s)
- Jingwen Hu
- Maternal-Fetal Medicine Institute, Department of Obstetrics and Gynaecology, Shenzhen Baoan Women’s and Children’s Hospital, Shenzhen University, Shenzhen, China
| | - Lin Chen
- Fertility Preservation Research Center, Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jing Ruan
- Maternal-Fetal Medicine Institute, Department of Obstetrics and Gynaecology, Shenzhen Baoan Women’s and Children’s Hospital, Shenzhen University, Shenzhen, China
| | - Xiaoyan Chen
- Maternal-Fetal Medicine Institute, Department of Obstetrics and Gynaecology, Shenzhen Baoan Women’s and Children’s Hospital, Shenzhen University, Shenzhen, China
- Fertility Preservation Research Center, Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
49
|
Thadhani R, Cerdeira AS, Karumanchi SA. Translation of mechanistic advances in preeclampsia to the clinic: Long and winding road. FASEB J 2024; 38:e23441. [PMID: 38300220 DOI: 10.1096/fj.202301808r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/30/2023] [Accepted: 01/09/2024] [Indexed: 02/02/2024]
Abstract
As one of the leading causes of premature birth and maternal and infant mortality worldwide, preeclampsia remains a major unmet public health challenge. Preeclampsia and related hypertensive disorders of pregnancy are estimated to cause >75 000 maternal and 500 000 infant deaths globally each year. Because of rising rates of risk factors such as obesity, in vitro fertilization and advanced maternal age, the incidence of preeclampsia is going up with rates ranging from 5% to 10% of all pregnancies worldwide. A major discovery in the field was the realization that the clinical phenotypes related to preeclampsia, such as hypertension, proteinuria, and other adverse maternal/fetal events, are due to excess circulating soluble fms-like tyrosine kinase-1 (sFlt-1, also referred to as sVEGFR-1). sFlt-1 is an endogenous anti-angiogenic protein that is made by the placenta and acts by neutralizing the pro-angiogenic proteins vascular endothelial growth factor (VEGF) and placental growth factor (PlGF). During the last decade, this work has spawned a new era of molecular diagnostics for early detection of this condition. Antagonizing sFlt-1 either by reducing production or blocking its actions has shown salutary effects in animal models. Further, in early-stage human studies, the therapeutic removal of sFlt-1 from maternal circulation has shown promise in delaying disease progression and improving outcomes. Recently, the FDA approved the first molecular test for preterm preeclampsia (sFlt-1/PlGF ratio) for clinical use in the United States. Measuring serum sFlt-1/PlGF ratio in the acute hospital setting may aid short-term management, particularly regarding step-up or step-down of care, decision to transfer to settings better equipped to manage both the mother and the preterm neonate, appropriate timing of administration of steroids and magnesium sulfate, and in expectant management decisions. The test itself has the potential to save lives. Furthermore, the availability of a molecular test that correlates with adverse outcomes has set the stage for interventional clinical trials testing treatments for this disorder. In this review, we will discuss the role of circulating sFlt-1 and related factors in the pathogenesis of preeclampsia and specifically how this discovery is leading to concrete advances in the care of women with preeclampsia.
Collapse
Affiliation(s)
- Ravi Thadhani
- Woodruff Health Sciences Center, Emory University School of Medicine, Atlanta, Georgia, USA
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Ana Sofia Cerdeira
- Nuffield Department of Women's Health and Reproductive Research, University of Oxford, Oxford, UK
- Fetal Maternal Medicine Unit, Queen Charlotte's and Chelsea Hospital, London, UK
| | - S Ananth Karumanchi
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
50
|
Kovacheva VP, Eberhard BW, Cohen RY, Maher M, Saxena R, Gray KJ. Preeclampsia Prediction Using Machine Learning and Polygenic Risk Scores From Clinical and Genetic Risk Factors in Early and Late Pregnancies. Hypertension 2024; 81:264-272. [PMID: 37901968 PMCID: PMC10842389 DOI: 10.1161/hypertensionaha.123.21053] [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: 02/07/2023] [Accepted: 10/12/2023] [Indexed: 10/31/2023]
Abstract
BACKGROUND Preeclampsia, a pregnancy-specific condition associated with new-onset hypertension after 20-weeks gestation, is a leading cause of maternal and neonatal morbidity and mortality. Predictive tools to understand which individuals are most at risk are needed. METHODS We identified a cohort of N=1125 pregnant individuals who delivered between May 2015 and May 2022 at Mass General Brigham Hospitals with available electronic health record data and linked genetic data. Using clinical electronic health record data and systolic blood pressure polygenic risk scores derived from a large genome-wide association study, we developed machine learning (XGBoost) and logistic regression models to predict preeclampsia risk. RESULTS Pregnant individuals with a systolic blood pressure polygenic risk score in the top quartile had higher blood pressures throughout pregnancy compared with patients within the lowest quartile systolic blood pressure polygenic risk score. In the first trimester, the most predictive model was XGBoost, with an area under the curve of 0.74. In late pregnancy, with data obtained up to the delivery admission, the best-performing model was XGBoost using clinical variables, which achieved an area under the curve of 0.91. Adding the systolic blood pressure polygenic risk score to the models did not improve the performance significantly based on De Long test comparing the area under the curve of models with and without the polygenic score. CONCLUSIONS Integrating clinical factors into predictive models can inform personalized preeclampsia risk and achieve higher predictive power than the current practice. In the future, personalized tools can be implemented to identify high-risk patients for preventative therapies and timely intervention to improve adverse maternal and neonatal outcomes.
Collapse
Affiliation(s)
- Vesela P Kovacheva
- Department of Anesthesiology, Perioperative and Pain Medicine (V.P.K., B.W.E., R.Y.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Braden W Eberhard
- Department of Anesthesiology, Perioperative and Pain Medicine (V.P.K., B.W.E., R.Y.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Raphael Y Cohen
- Department of Anesthesiology, Perioperative and Pain Medicine (V.P.K., B.W.E., R.Y.C.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- PathAI, Boston, MA (R.Y.C.)
| | - Matthew Maher
- Department of Anesthesia, Critical Care and Pain Medicine, Center for Genomic Medicine, Massachusetts General Hospital, Boston (M.M., R.S., K.J.G.)
| | - Richa Saxena
- Department of Anesthesia, Critical Care and Pain Medicine, Center for Genomic Medicine, Massachusetts General Hospital, Boston (M.M., R.S., K.J.G.)
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston (R.S.)
| | - Kathryn J Gray
- Division of Maternal-Fetal Medicine (K.J.G.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Department of Anesthesia, Critical Care and Pain Medicine, Center for Genomic Medicine, Massachusetts General Hospital, Boston (M.M., R.S., K.J.G.)
| |
Collapse
|