1
|
Lee DI, Roy S. Examining the dynamics of three-dimensional genome organization with multitask matrix factorization. Genome Res 2025; 35:1179-1193. [PMID: 40113262 PMCID: PMC12047540 DOI: 10.1101/gr.279930.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
Three-dimensional (3D) genome organization, which determines how the DNA is packaged inside the nucleus, has emerged as a key component of the gene regulation machinery. High-throughput chromosome conformation data sets, such as Hi-C, have become available across multiple conditions and time points, offering a unique opportunity to examine changes in 3D genome organization and link them to phenotypic changes in normal and disease processes. However, systematic detection of higher-order structural changes across multiple Hi-C data sets remains a major challenge. Existing computational methods either do not model higher-order structural units or cannot model dynamics across more than two conditions of interest. We address these limitations with tree-guided integrated factorization (TGIF), a generalizable multitask nonnegative matrix factorization (NMF) approach that can be applied to time series or hierarchically related biological conditions. TGIF can identify large-scale changes at the compartment or subcompartment levels, as well as local changes at boundaries of topologically associated domains (TADs). Based on benchmarking in simulated and real Hi-C data, TGIF boundaries are more accurate and reproducible across differential levels of noise and sources of technical artifacts, and are more enriched in CTCF. Application to three multisample mammalian data sets shows that TGIF can detect differential regions at compartment, subcompartment, and boundary levels that are associated with significant changes in regulatory signals and gene expression enriched in tissue-specific processes. Finally, we leverage TGIF boundaries to prioritize sequence variants for multiple phenotypes from the NHGRI GWAS catalog. Taken together, TGIF is a flexible tool to examine 3D genome organization dynamics across disease and developmental processes.
Collapse
Affiliation(s)
- Da-Inn Lee
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin 53715, USA
| | - Sushmita Roy
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin 53715, USA;
- Wisconsin Institute for Discovery, Madison, Wisconsin 53715, USA
| |
Collapse
|
2
|
Lanting V, Oskam M, Wilmink H, Kamphuisen PW, van Es N. The role of germline and somatic mutations in predicting cancer-associated thrombosis: a narrative review. Curr Opin Hematol 2025; 32:138-145. [PMID: 39851266 PMCID: PMC11957438 DOI: 10.1097/moh.0000000000000861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
PURPOSE OF REVIEW Patients with cancer have an increased risk of venous thromboembolism (VTE). Guidelines suggest to use risk assessment tools to guide decisions about thromboprophylaxis, but current tools have modest discriminatory ability. Genetic information from the germline or tumor has the potential to improve VTE prediction. Here, we provide a clinical overview of the current role of genetics in cancer-associated VTE. RECENT FINDINGS Germline mutations, such as factor V Leiden and prothrombin G20210A, are associated with a 2- to 2.5-fold increased VTE risk in patients with cancer. Tumor-specific somatic mutations also contribute to VTE risk, such as ALK rearrangements increasing the risk in nonsmall cell lung cancer and IDH1 mutations decreasing the risk in gliomas. Other somatic mutations associated with VTE independent of tumor type include KRAS , STK11 , MET , KEAP1 , CTNNB1 , and CDKN2B . Incorporating data on germline or somatic mutations in risk scores improves discriminatory ability compared with the Khorana score. SUMMARY Specific germline and somatic mutations are associated with an increased VTE risk in patients with cancer and potentially improve performance of clinical risk scores. The increasing and widespread use of genetic testing in cancer care provides an opportunity for further development of prediction models incorporating genetic predictors.
Collapse
Affiliation(s)
- Vincent Lanting
- Amsterdam UMC, University of Amsterdam, department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam
- Tergooi MC, department of Internal Medicine, Hilversum
| | - Merel Oskam
- Amsterdam UMC, University of Amsterdam, department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam
| | - Hanneke Wilmink
- Amsterdam UMC, University of Amsterdam, department of Oncology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Pieter W. Kamphuisen
- Amsterdam UMC, University of Amsterdam, department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam
- Tergooi MC, department of Internal Medicine, Hilversum
| | - Nick van Es
- Amsterdam UMC, University of Amsterdam, department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam
| |
Collapse
|
3
|
Haysen SR, Nielsen ALL, Qvist P, Kragstrup TW. Investigating associations between JAK inhibition and venous thromboembolism by systematic mining of large-scale datasets. Inflammopharmacology 2025; 33:1425-1434. [PMID: 39994070 PMCID: PMC11913929 DOI: 10.1007/s10787-025-01677-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/31/2025] [Indexed: 02/26/2025]
Abstract
Janus kinase inhibitors (JAKi) have been associated with an increased risk of venous thromboembolism (VTE) limiting the use of JAKi-based therapy. To improve risk stratification and drug development, it is crucial to understand the implication of dysregulated JAK-Signal Transducers and Activators of Transcription (STAT) signaling in the pathogenesis of VTE. The objective of this study is to clarify the putative genomic vulnerability to dysregulated JAK-STAT signaling in VTE through systematic mining of large-scale datasets generated from studies comparing VTE patients with healthy controls. Particularly, we assess the representation of entities of the JAK-STAT signaling pathway including STAT target genes among sets of miRNA, mRNA, and proteins differentially abundant in VTE patients, and we explore the putative cumulative genetic association of JAK-STAT signaling gene sets to VTE. Genes related to the JAK-STAT pathway were found significantly altered in VTE patients compared to healthy controls, indicating that genes under transcriptional control of STAT may be dysregulated in VTE. In support of this notion, we find a significant overrepresentation of predicted STAT target genes among genes downregulated in VTE patients, and promoter sequences of differentially regulated genes were significantly enriched with STAT transcription factor binding site motifs. Further linking STAT signaling to the molecular signature of VTE, genes targeted by miRNAs differentially regulated in patients are significantly enriched with STAT target genes and genes acting in the JAK-STAT signaling pathway. Together, our findings indicate that disruptions in the JAK-STAT pathway contribute to the molecular profile of VTE. This offers hope for identifying ways to interact with the JAK-STAT pathway that do not carry the risk of VTE.
Collapse
Affiliation(s)
| | | | - Per Qvist
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Centre for Genomics and Personalized Medicine, CGPM, Aarhus University, Aarhus, Denmark
| | - Tue Wenzel Kragstrup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
- Rheumatology Sector, Medical Diagnostic Center, Silkeborg Regional Hospital, Silkeborg, Denmark.
| |
Collapse
|
4
|
Pelland-Marcotte MC, Belaktib A, Droit A, Remy MM, Clement JG, Bianco S, Ma Y, Liu J, Herrmann L, Raufaste-Cazavieille V, Joly-Beauparlant C, Mangnier L, Leclercq M, Sontag T, Caron M, St-Onge P, Langlois S, Koch V, Flamand Y, Sinnett D, Silverman L, Tran TH, Santiago R. Molecular signatures associated with venous thromboembolism in children with acute lymphoblastic leukemia. J Thromb Haemost 2025; 23:1009-1022. [PMID: 39694444 DOI: 10.1016/j.jtha.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/14/2024] [Accepted: 12/01/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Venous thromboembolism (VTE) is a frequent complication of childhood acute lymphoblastic leukemia (ALL). OBJECTIVES We aimed to identify molecular markers and signatures of the leukemia microenvironment associated with VTE in childhood ALL by the dual-omics approach of gene expression and DNA methylation profiling. METHODS Eligible children aged 1 to 21 years old with newly diagnosed ALL were enrolled in the Dana-Farber Cancer Institute 16-001 trial with available RNA sequencing data from bone marrow at diagnosis. The primary outcome was VTE requiring medical intervention, divided between early events (ETs), within 6 weeks from ALL diagnosis, or late events otherwise. We compared differential gene expression and DNA methylation in children with and without VTE and in the subgroup of children with ETs. The DNA methylation cis-regulation was explored by dual-omics integration. Functional gene set enrichment analyses were performed to assess dysregulated pathways associated with thrombosis. Gene expression profiling-based signature for the thrombosis-free interval was determined using the Kaplan-Meier estimator and log-rank tests. RESULTS We included 248 patients (median age, 7.5 years; 78% precursor B-cell ALL), of whom 56 (23%) developed VTE. Genes and metabolic pathways involved in coagulation, platelet activation, and neutrophil extracellular trap formation were associated with ETs. Dual-omics analysis indicated that methylation reprogramming might be responsible for the overexpression of genes involved in neutrophil extracellular trap formation and coagulation in patients with ETs. A prothrombotic gene signature, based on VWF, PF4, and CXCL8 expression, predicted a thrombosis-free interval. CONCLUSION This suggests that gene markers and epigenetic regulation of the leukemic microenvironment are drivers of VTE, notably ETs in childhood ALL.
Collapse
Affiliation(s)
- Marie-Claude Pelland-Marcotte
- Department of Pediatrics, Centre Hospitalier Universitaire de Québec - Centre Mère-Enfant Soleil, Quebec City, Quebec, Canada; Centre de Recherche du Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada
| | - Anas Belaktib
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada
| | - Arnaud Droit
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada
| | | | - Jeyani George Clement
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada; Department of Immunity and Cancer, Institut Curie, Paris, France
| | - Stéphanie Bianco
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada
| | - Yan Ma
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada
| | - Jessica Liu
- Department of Pediatrics, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Lara Herrmann
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada
| | | | | | - Loïc Mangnier
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada
| | - Mickael Leclercq
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada
| | - Thomas Sontag
- Axis of Immune Diseases and Cancers, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, Quebec, Canada
| | - Maxime Caron
- Axis of Immune Diseases and Cancers, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, Quebec, Canada
| | - Pascal St-Onge
- Axis of Immune Diseases and Cancers, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, Quebec, Canada
| | - Sylvie Langlois
- Axis of Immune Diseases and Cancers, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, Quebec, Canada
| | - Victoria Koch
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, Massachusetts, USA
| | - Yael Flamand
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Daniel Sinnett
- Axis of Immune Diseases and Cancers, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, Quebec, Canada; Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, Quebec, Canada
| | - Lewis Silverman
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Thai Hoa Tran
- Axis of Immune Diseases and Cancers, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, Quebec, Canada; Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, Quebec, Canada
| | - Raoul Santiago
- Department of Pediatrics, Centre Hospitalier Universitaire de Québec - Centre Mère-Enfant Soleil, Quebec City, Quebec, Canada; Centre de Recherche du Centre Hospitalier Universitaire de Québec, Quebec City, Quebec, Canada.
| |
Collapse
|
5
|
Meng Z, Zhang C, Liu S, Li W, Wang Y, Zhang Q, Peng B, Ye W, Jiang Y, Song Y, Guo M, Chang X, Shao L. Exploring genetic loci linked to COVID-19 severity and immune response through multi-trait GWAS analyses. Front Genet 2025; 16:1502839. [PMID: 40034745 PMCID: PMC11873281 DOI: 10.3389/fgene.2025.1502839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction COVID-19 severity has been linked to immune factors, with excessive immune responses like cytokine storms contributing to mortality. However, the genetic basis of these immune responses is not well understood. This study aimed to explore the genetic connection between COVID-19 severity and blood cell traits, given their close relationship with immunity. Materials and methods GWAS summary statistics for COVID-19 and blood cell counts were analyzed using Linkage Disequilibrium Score Regression (LDSC) to estimate genetic correlations and heritabilities. For traits with significant correlations, a Multi-Trait GWAS Analysis (MTAG) was performed to identify pleiotropic loci shared between COVID-19 and blood cell counts. Results Our MTAG analysis identified four pleiotropic loci associated with COVID-19 severity, five loci linked to hospitalized cases, and one locus related to general patients. Among these, two novel loci were identified in the high-risk population, with rs55779981 located near RAVER1 and rs73009538 near CARM1. In hospitalized patients, two previously unrecognized loci were detected, namely, rs115545251 near GFI1 and rs3181049 near RAVER1, while in general patients, rs11065822 near CUX2 emerged as a newly identified locus. We also identified potential target genes, including those involved in inflammation signaling (CARM1), endothelial dysfunction (INTS12), and antiviral immune response (RAVER1), which may require further investigation. Conclusion Our study offers insights into the genetic overlap between COVID-19 and immune factors, suggesting potential directions for future research and clinical exploration.
Collapse
Affiliation(s)
- Ziang Meng
- Department of Infectious Disease, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Chumeng Zhang
- The Second School of Clinical Medicine of Shandong First Medical University, Tai’an, Shandong, China
| | - Shuai Liu
- Agricultural Products Quality and Safety Center of Jinan, Jinan, Shandong, China
| | - Wen Li
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Yue Wang
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Qingyi Zhang
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Bichen Peng
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Weiyi Ye
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Yue Jiang
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Yingchao Song
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Miao Guo
- School of Life Sciences, Shandong First Medical University, Shandong, China
| | - Xiao Chang
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Lei Shao
- Department of Infectious Disease, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
6
|
Bankier S, Gudmundsdottir V, Jonmundsson T, Bjarnadottir H, Loureiro J, Wang L, Finkel N, Orth AP, Aspelund T, Launer LJ, Björkegren JL, Jennings LL, Lamb JR, Gudnason V, Michoel T, Emilsson V. Circulating causal protein networks linked to future risk of myocardial infarction. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.07.25321789. [PMID: 39974043 PMCID: PMC11838656 DOI: 10.1101/2025.02.07.25321789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Variations in blood protein levels have been associated with a broad spectrum of complex diseases, including atherosclerotic cardiovascular disease (ACVD). These associations highlight the intricate interplay between local (e.g., cardiovascular) and systemic (non-cardiovascular) factors for the development of ACVD, emphasizing the need for a comprehensive, systems-level understanding of its etiology. To accomplish this, we developed a causal network inference framework by analyzing one of the largest serum proteomics studies to date, the Age, Gene/Environment Susceptibility-Reykjavik Study (AGES), a prospective population-based study of 7,523 serum proteins measured in 5,376 older adults. To reconstruct a causal network of serum proteins, we used cis -acting protein quantitative trait loci (pQTLs) as instrumental variables to infer causal relationships between protein pairs, while accounting for potential unobserved confounding factors. We identified 185 causal protein subnetworks (FDR = 1%, n ≥ 10 members), which collectively interacted with 5,611 target proteins, offering valuable biological insights and an overview of systemic homeostasis. Several subnetworks, many of which interact to establish a hierarchy of directional relationships, were significantly associated with future myocardial infarction and/or its long-term complications like heart failure, as well as with key cardiometabolic traits that contribute to the onset of ACVD.
Collapse
|
7
|
Memon AA, Zöller B, Svensson PJ, Sundquist J, Sundquist K. Fibrinogen genotypes and their impact on recurrence of venous thromboembolism and family history: A prospective population-based study. Br J Haematol 2025; 206:657-665. [PMID: 39828282 PMCID: PMC11829138 DOI: 10.1111/bjh.19999] [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: 10/28/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Venous thromboembolism (VTE) involves blood clot formation in veins, resulting in serious health issues. Fibrinogen, a crucial clotting protein, consists of three polypeptides encoded by the fibrinogen genes: alpha (FGA), beta (FGB) and gamma (FGG). We genotyped most common missense variants in the fibrinogen genes in relation to VTE, recurrence and family history in Malmö Thrombophilia Study, including 1465 VTE patients followed for ~10 years and 429 healthy donors. FGG (rs6063) was significantly associated with increased odds of primary VTE (odds ratio [OR] = 8.2; 95% confidence interval [CI] = 1.05-63.6) after adjusting for age and sex. For recurrent VTE, Cox-regression analysis indicated a higher risk associated with FGA (rs6050) (hazard ratio [HR] = 1.8; 95% CI = 1.1-2.8), with even greater risk for unprovoked recurrent VTE (HR = 2.3; 95% CI = 1.3-4.2), surpassing the well-known factor V Leiden (FVL) (HR = 1.9; 95% CI = 1.2-3.0). Combining risk alleles from FVL and FGA (rs6050) significantly raised the risk for unprovoked recurrent VTE: ≥3 risk alleles (HR = 4.6; 95% CI = 1.9-11.3), two risk alleles (HR = 2.6; 95% CI = 1.4-4.8) and one risk allele (HR = 1.5; 95% CI = 0.8-2.7) compared to 0 risk allele. Prevalence of FGA (rs6050) risk allele was significantly higher in cases with a family history of VTE. We propose FGA (rs6050) as a novel predictor for unprovoked recurrent VTE and it may contribute to the familial occurrence of VTE.
Collapse
Affiliation(s)
| | - Bengt Zöller
- Center for Primary Care ResearchLund UniversityMalmoSweden
| | | | - Jan Sundquist
- Center for Primary Care ResearchLund UniversityMalmoSweden
| | | |
Collapse
|
8
|
Li S, Jiang M, Guan Y, Cao X, Shao Z, Deng J, Hao X. Association between genetic risk and adherence to the Dietary Approaches to Stop Hypertension diet for developing venous thromboembolism. Res Pract Thromb Haemost 2025; 9:102731. [PMID: 40236289 PMCID: PMC11999685 DOI: 10.1016/j.rpth.2025.102731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 01/23/2025] [Accepted: 02/25/2025] [Indexed: 04/17/2025] Open
Abstract
Background The relationship between diet and venous thromboembolism (VTE) remains unclear, and the joint effects of diet patterns and genetic susceptibility on VTE risk are unknown. Objectives Investigate the independent and joint effects of Dietary Approaches to Stop Hypertension (DASH) diet adherence and polygenic risk scores (PRS) on VTE risk. Methods A total of 411,539 UK Biobank participants were included. DASH scores were calculated using Food Frequency Questionnaires, and PRS quantified genetic risk. Cox proportional hazard models estimated hazard ratios (HRs) for VTE, assessing interactions between the DASH diet and genetic susceptibility. Results During a median follow-up of 13.4 years, 10,543 participants were diagnosed with VTE. Higher DASH scores were associated with a lower VTE risk (HR, 0.87; 95% CI, 0.82-0.92). A low-adherent DASH diet combined with high-genetic risk had the highest VTE risk (HR, 2.78; 95% CI, 2.47-3.14). High DASH scores reduced VTE risk in high-genetic-risk individuals (HR, 0.84; 95% CI, 0.76-0.92). Sex-specific associations were detected in the joint effect and interaction of DASH scores and PRS. Notably, high DASH scores can offset moderate genetic risk among men (HR, 0.79; 95% CI, 0.67-0.94). There were additive interactions between DASH scores and high genetic risk in total subjects and men, while not observed in women. Conclusion The DASH diet is associated with reduced VTE risk and can partially offset genetic predisposition. Low adherence to the DASH diet increases VTE risk, particularly in high-genetic-risk individuals. The protective effect of high DASH scores against genetic risks for VTE is more pronounced in males. Precision medicine should consider both diet and genetics for VTE prevention.
Collapse
Affiliation(s)
- Si Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Minghui Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yunlong Guan
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xi Cao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhonghe Shao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Deng
- Institute of Hematology, Union Hospital, Tongii Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
9
|
Jiang Z, Sullivan PF, Li T, Zhao B, Wang X, Luo T, Huang S, Guan PY, Chen J, Yang Y, Stein JL, Li Y, Liu D, Sun L, Zhu H. The X chromosome's influences on the human brain. SCIENCE ADVANCES 2025; 11:eadq5360. [PMID: 39854466 PMCID: PMC11759047 DOI: 10.1126/sciadv.adq5360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025]
Abstract
Genes on the X chromosome are extensively expressed in the human brain. However, little is known for the X chromosome's impact on the brain anatomy, microstructure, and functional networks. We examined 1045 complex brain imaging traits from 38,529 participants in the UK Biobank. We unveiled potential autosome-X chromosome interactions while proposing an atlas outlining dosage compensation for brain imaging traits. Through extensive association studies, we identified 72 genome-wide significant trait-locus pairs (including 29 new associations) that share genetic architectures with brain-related disorders, notably schizophrenia. Furthermore, we found unique sex-specific associations and assessed variations in genetic effects between sexes. Our research offers critical insights into the X chromosome's role in the human brain, underscoring its contribution to the differences observed in brain structure and functionality between sexes.
Collapse
Affiliation(s)
- Zhiwen Jiang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Patrick F. Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tengfei Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xifeng Wang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tianyou Luo
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Shuai Huang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Peter Y. Guan
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jie Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yue Yang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jason L. Stein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dajiang Liu
- Department of Public Health Sciences, Penn State University, Hershey, PA 17033, USA
- Department of Biochemistry and Molecular Biology, Penn State University, Hershey, PA 17033, USA
| | - Lei Sun
- Department of Statistical Sciences, University of Toronto, Toronto, ON M5G 1Z5, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
10
|
Zöller B, Sundquist J, Sundquist K, Ohlsson H. The risk for psychiatric disorders in offspring from thrombosis-prone pedigrees in Sweden: a nationwide family study. Res Pract Thromb Haemost 2025; 9:102692. [PMID: 40093964 PMCID: PMC11908565 DOI: 10.1016/j.rpth.2025.102692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/14/2024] [Accepted: 01/22/2025] [Indexed: 03/19/2025] Open
Abstract
Background Psychiatric disorders have been associated with venous thromboembolism (VTE). However, to our knowledge, no nationwide study has examined the familial association between VTE and psychiatric disorders. Objectives We took a pedigree-based approach and examined the risk of psychiatric disorders in offspring from extended pedigrees according to the densities of VTE in pedigrees. Methods This was a Swedish national family study. We identified a total of 482,184 Swedish pedigrees from the Swedish Multigeneration Register containing a mean of 14.2 parents, aunts/uncles, grandparents, and cousins of a core full-sibship that we termed the pedigree offspring (n = 751,060). We then derived 8 empirical classes of these pedigrees based on the density of cases of VTE. The risk was determined in offspring for psychiatric disorders as a function of VTE density in their pedigrees. Diagnoses of VTE and psychiatric disorders (F00-F69) were determined according to the International Classification of Diseases codes in Swedish registers. All results were Bonferroni corrected. Results Higher VTE density, especially for females in pedigrees, was significantly but weakly associated in the offspring with a higher risk of psychiatric disorders. Moreover, VTE density in pedigrees was significantly associated in the offspring with substance abuse disorders, mood (affective) disorders, neurotic, stress-related, and somatoform disorders, behavioral syndromes associated with psychological disturbances and physical factors, personality disorders of adult personality and behavior, depression, and anxiety disorders. Conclusion Offspring of pedigrees with a high density of VTE, especially for females, are slightly disadvantaged regarding several psychiatric disorders. VTE shares familial susceptibility, albeit weak, with several psychiatric disorders.
Collapse
Affiliation(s)
- Bengt Zöller
- Department of Clinical Sciences, Malmö, Center for Primary Health Care Research, Lund University, Region Skåne, Malmö, Sweden
| | - Jan Sundquist
- Department of Clinical Sciences, Malmö, Center for Primary Health Care Research, Lund University, Region Skåne, Malmö, Sweden
| | - Kristina Sundquist
- Department of Clinical Sciences, Malmö, Center for Primary Health Care Research, Lund University, Region Skåne, Malmö, Sweden
| | - Henrik Ohlsson
- Department of Clinical Sciences, Malmö, Center for Primary Health Care Research, Lund University, Region Skåne, Malmö, Sweden
| |
Collapse
|
11
|
Smeets MJ, Petersen PB, Jørgensen CC, Cannegieter SC, Ostrowski SR, Kehlet H, Nemeth B. Validation of the 5-SNP score for the prediction of venous thromboembolism in a Danish fast-track cohort of 6789 total hip and total knee arthroplasty patients. Res Pract Thromb Haemost 2025; 9:102644. [PMID: 39810985 PMCID: PMC11731481 DOI: 10.1016/j.rpth.2024.102644] [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: 07/26/2024] [Revised: 11/08/2024] [Accepted: 11/21/2024] [Indexed: 01/16/2025] Open
Abstract
Background Venous thromboembolism (VTE) is a serious complication following total hip arthroplasty (THA) and total knee arthroplasty (TKA). Despite improvements with fast-track treatment protocols, 0.5% of patients still develop a VTE within 90-days postoperatively. Previously, the 5-single nucleotide polymorphism (SNP) genetic risk scores (weighted and simplified) were developed to identify people at a high risk for VTE within the general population. Objectives We aimed to assess whether the 5-SNP scores could be used to identify high-risk patients in a cohort of fast-track THA/TKA patients. Methods A subset of patients from the Lundbeck Centre for Fast-track Hip and Knee Replacement Database was included based on the availability of genetic information. The 5-SNP scores were calculated for these patients, and their discriminatory performance was determined by c-statistic. Furthermore, the 5-SNP scores were added to a simple logistic prediction model containing clinical predictors to assess the added predictive value. Results A total of 7753 THA and TKA procedures (6798 patients) were included in this study. The c-statistics for the weighted and simple 5-SNP scores were 0.50 (95% CI, 0.39-0.61) and 0.48 (95% CI, 0.38-0.58), respectively. For the model with clinical predictors, the c-statistic was 0.67 (95% CI, 0.56-0.77). Addition of either of the 5-SNP scores did not improve discrimination in this model. Conclusion These findings do not support genetic risk profiling in fast-track THA/TKA patients to predict VTE. Hence, efforts should be directed at optimizing prediction models with clinical predictors.
Collapse
Affiliation(s)
- Mark J.R. Smeets
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Pelle B. Petersen
- Section of Surgical Pathophysiology, 7621, Rigshospitalet, Copenhagen, Denmark
| | - Christoffer C. Jørgensen
- The Lundbeck Foundation Centre for Fast-Track Hip and Knee Replacement, 7621, Rigshospitalet, Copenhagen, Denmark
| | - Suzanne C. Cannegieter
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Internal Medicine - Section Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Sisse R. Ostrowski
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Henrik Kehlet
- Section of Surgical Pathophysiology, 7621, Rigshospitalet, Copenhagen, Denmark
- The Lundbeck Foundation Centre for Fast-Track Hip and Knee Replacement, 7621, Rigshospitalet, Copenhagen, Denmark
| | - Banne Nemeth
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Orthopaedic Surgery, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
12
|
Wang J, Xie Y, Zeng C, Nie L, Yang Y. Predictive value of abnormal expression of MPHOSPH9 in reintervention after high intensity focused ultrasound treatment of uterine fibroids. J Obstet Gynaecol Res 2025; 51:e16205. [PMID: 39780408 DOI: 10.1111/jog.16205] [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: 08/27/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025]
Abstract
AIMS Aberrantly expressed MPHOSPH9 has been reported to be associated with poor prognosis in many diseases. Previous study indicates that MPHOSPH9 is abnormally expressed in patients with uterine fibroids (UFs). This study focused on the possible prognostic value of MPHOSPH9 in UFs patients after high intensity focused ultrasound (HIFU) treatment. METHODS A total of 455 UFs patients participated in the study, including 95 patients who needed reintervention (Reintervention group) and 360 patients who did not need reintervention (Non-reintervention group) after HIFU treatment. They volunteered blood samples before HIFU treatment. The relative expression of MPHOSPH9 was assessed by qRT-PCR. Cox regression analysis was performed to assess the prognostic value of MPHOSPH9 and risk factors of postoperative reintervention. RESULTS The expression of MPHOSPH9 was markedly elevated in Reintervention, compared to that of the Non-reintervention group (p < 0.001). Five-year follow-up results showed that among these postoperative patients, 95 patients required reintervention and the reintervention time mainly occurred during 10-40 months after surgery. The Cox regression analysis demonstrated that UFs size (hazard ratio = 2.769, 95% confidence interval: 1.366-5.610) and the high MPHOSPH9 expression level (hazard ratio = 3.160, 95% confidence interval: 1.506-6.630) were independent risk factors for postoperative reintervention. CONCLUSIONS The enhanced MPHOSPH9 was a potential candidate for predicting postoperative reintervention in UFs patients.
Collapse
Affiliation(s)
- Jing Wang
- Department of Imaging, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, Nanchong, China
| | - Yingjie Xie
- One Departments of Cadre Ward, General Hospital of Southern Theater Command of Chinese PLA, Guangzhou, China
| | - Chaoqiang Zeng
- Department of Imaging, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, Nanchong, China
| | - Lin Nie
- Department of Imaging, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, Nanchong, China
| | - Yang Yang
- Department of Radiology, Ya'an People's Hospital, Ya'an, China
| |
Collapse
|
13
|
Kvasnicka T, Cifkova R, Zenahlikova Z, Bobcikova P, Syruckova A, Sevcik M, Dusková D, Kvasnicka J. The Prevalence of the Thrombotic SNPs rs6025, rs1799963, rs2066865, rs2289252 and rs8176719 in Patients with Venous Thromboembolism in the Czech Population. Clin Appl Thromb Hemost 2025; 31:10760296251324202. [PMID: 40094632 PMCID: PMC11915282 DOI: 10.1177/10760296251324202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025] Open
Abstract
IntroductionStudy aimed to determine the occurrence of 5 thrombosis-related single-nucleotide polymorphisms (SNPs) in patients with venous thromboembolism (VTE) (n = 2630) and a control group (n = 2637) in the Czech population.MethodsThe following gene SNPs were detected in both groups: F5 Leiden (rs6025), F2 (rs1799963), FGG, fibrinogen gamma' (rs2066865), F11 (rs2289252) and ABO (rs8176719). Statistical analysis was performed using SAS statistical software with population genetics tools.ResultsHeterozygotes for F5 Leiden were associated with a 5.58-fold and homozygotes F5 Leiden with a 33.46-fold increased risk of VTE. At SNP rs1799963 (F2, prothrombin), only heterozygotes had a significant 3.9-fold increased risk of VTE. The findings at SNP rs2066865 (fibrinogen gamma', FGG) showed a 1.37-fold increased risk of VTE for FGG heterozygotes and a 1.77-fold increased risk of VTE for FGG homozygotes. There is also a significant 1.42-fold increase risk of VTE in the heterozygotes and a 1.80-fold increase risk of VTE in the homozygotes of the SNP rs 2289252 (F11). Further higher increases in the risk of VTE in both variants were found in patients with VTE at rs8176719 (ABO, non-O). It corresponds to a 2.2-fold increase in the risk of VTE in heterozygotes and a 3.5-fold increase in the risk of VTE in homozygotes.ConclusionBesides F5 Leiden and prothrombin mutation, the study suggests that the gene polymorphisms of FGG (rs2066865), F11 (rs2289252) and ABO (rs8176719) play a role as an independent heritable risk factor for VTE in the Czech population.
Collapse
Affiliation(s)
- Tomas Kvasnicka
- Thrombotic Centre, Institute of Medical Biochemistry and Laboratory Diagnostics, first Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Renata Cifkova
- Center for Cardiovascular Prevention, first Faculty of Medicine, Charles University and Thomayer University Hospital, Prague, Czech Republic
| | - Zuzana Zenahlikova
- Thrombotic Centre, Institute of Medical Biochemistry and Laboratory Diagnostics, first Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Petra Bobcikova
- Thrombotic Centre, Institute of Medical Biochemistry and Laboratory Diagnostics, first Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Alena Syruckova
- Thrombotic Centre, Institute of Medical Biochemistry and Laboratory Diagnostics, first Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Martin Sevcik
- Thrombotic Centre, Institute of Medical Biochemistry and Laboratory Diagnostics, first Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Daniela Dusková
- 1st Department of Medicine, Division of Hematology, first Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- Department of Blood Transfusion, first Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jan Kvasnicka
- 1st Department of Medicine, Division of Hematology, first Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| |
Collapse
|
14
|
Vostatek R, Ay C. Biological Aging and Venous Thromboembolism: A Review of Telomeres and Beyond. Biomedicines 2024; 13:15. [PMID: 39857599 PMCID: PMC11759860 DOI: 10.3390/biomedicines13010015] [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: 11/09/2024] [Revised: 11/29/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025] Open
Abstract
Although venous thromboembolism (VTE) is the third most common cardiovascular disease, and the risk of VTE increases sharply with advancing age, approximately 40% of VTE cases are currently classified as unprovoked, highlighting the importance of risk factor research. While chronological aging is associated with the risk of VTE, the association with biological aging remains unclear. Biological aging is highly complex, influenced by several dysregulated cellular and biochemical mechanisms. In the last decade, advancements in omics methodologies provided insights into the molecular complexity of biological aging. Techniques such as high-throughput genomics, epigenomics, transcriptomics, proteomics, and metabolomics analyses identified and quantified numerous epigenetic markers, transcripts, proteins, and metabolites. These methods have also revealed the molecular alterations organisms undergo as they age. Despite the progress, there is still a lack of consensus regarding the methods for assessing and validating these biomarkers, and their application lacks standardization. This review gives an overview of biomarkers of biological aging, including telomere length, and their potential role for VTE. Furthermore, we critically examine the advantages and disadvantages of the proposed methods and discuss possible future directions for investigating biological aging in VTE.
Collapse
Affiliation(s)
| | - Cihan Ay
- Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
15
|
Xia YQ, Tang L, Hu Y. [Advances in the genetics of venous thromboembolic disease]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2024; 45:1144-1147. [PMID: 39765359 PMCID: PMC11886693 DOI: 10.3760/cma.j.cn121090-20240327-00117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Indexed: 03/09/2025]
Abstract
Venous thromboembolism (VTE) is clinically manifested as deep vein thrombosis (DVT) and pulmonary embolism (PE). VTE is the third most prevalent vascular disease after coronary artery and cerebrovascular diseases. VTE is a multifactorial disease caused by the interaction of genetic and acquired risk factors. Genetic heritability is estimated to be 40%-60% based on studies of families, twins, and siblings. The accumulation of information linking genetic variations to VTE risk has rapidly expanded with the continuous advancement of sequencing technology. Currently, mutations in key genes of the coagulation system, anticoagulation system, and fibrinolysis system are constantly being updated, and the functional mechanisms of new genes are receiving gaining attention. This review summarizes the research progress and prospects of key genetic variations associated with venous thromboembolism.
Collapse
Affiliation(s)
- Y Q Xia
- Institute of Hematology, Tongji Medical College Affiliated Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - L Tang
- Institute of Hematology, Tongji Medical College Affiliated Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Y Hu
- Institute of Hematology, Tongji Medical College Affiliated Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
16
|
Srivastava S, Garg I. Thrombotic complications post liver transplantation: Etiology and management. World J Crit Care Med 2024; 13:96074. [PMID: 39655303 PMCID: PMC11577539 DOI: 10.5492/wjccm.v13.i4.96074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 10/01/2024] [Accepted: 10/24/2024] [Indexed: 10/31/2024] Open
Abstract
Liver transplantation (LT) is the life saving therapeutic option for patients with acute and chronic end stage liver disease. This is a routine procedure with excellent outcomes in terms of patient survival and quality of life post LT. Orthotopic LT (OLT) patients require a critical care as they are prone to variety of post-operative vascular, cardiovascular, biliary, pulmonary and abdominal complications. Thrombotic complications (both arterial and venous) are not uncommon post liver transplant surgery. Such vascular problems lead to increased morbidity and mortality in both donor and graft recipient. Although thromboprophylaxis is recommended in general surgery patients, no such standards exist for liver transplant patients. Drastic advancements of surgical and anesthetic procedures have improvised survival rates of patients post OLT. Despite these, haemostatic imbalance leading to thrombotic events post OLT cause significant graft loss and morbidity and even lead to patient's death. Thus it is extremely important to understand pathophysiology of thrombosis in LT patients and shorten the timing of its diagnosis to avoid morbidity and mortality in both donor and graft recipient. Present review summarizes the current knowledge of vascular complications associated with LT to highlight their impact on short and long-term morbidity and mortality post LT. Also, present report discusses the lacunae existing in the literature regarding the risk factors leading to arterial and venous thrombosis in LT patients.
Collapse
Affiliation(s)
- Swati Srivastava
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development organization, Delhi 110054, India
| | - Iti Garg
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development organization, Delhi 110054, India
| |
Collapse
|
17
|
Bai J, Yang Z, Jia Y, Yu J, Jiang W, Liu Y, Li F, Zeng R, Wan Z, Lei Y, Liao X, Li D, Zhao Q. Sleep patterns, genetic susceptibility, and venous thromboembolism: A prospective study of 384,758 UK Biobank participants. PLoS One 2024; 19:e0309870. [PMID: 39240854 PMCID: PMC11379228 DOI: 10.1371/journal.pone.0309870] [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: 01/20/2024] [Accepted: 08/15/2024] [Indexed: 09/08/2024] Open
Abstract
BACKGROUND Although healthy sleep patterns have been linked to a lower risk of cardiovascular disease in earlier research, it is unclear how beneficial they are for venous thromboembolism (VTE). AIM This research aimed to examine the correlation between sleep patterns, genetic susceptibility, and VTE. METHODS In the UK Biobank cohort, healthy sleep behaviors were defined as early chronotype, 7-8 hours of sleep each day, no snoring, infrequent insomnia, and infrequent daytime sleepiness. Each of the five criteria was given 1 point, creating a healthy sleep score ranging from 0 to 5. Cox proportional hazards regression models were utilized to examine the associations between genetic susceptibility, healthy sleep score and VTE. RESULTS The UK Biobank study included 384,758 participants aged 56.6 ± 8.0 years. After a median of 11.9 years of follow-up, 8,885 (2.3%) participants were diagnosed with VTE. A healthy sleep score inversely affected VTE risk. For participants with a score of 5, the hazard ratio of VTE was 0.813 (95% confidence interval: 0.758-0.873, P<0.001) compared to those with a score ≤2. Early chronotype, sleeping 7-8 hours each day, infrequent insomnia, and infrequent daytime sleepiness were significantly associated with a 7.9%, 8.3%, 5.1%, and 20.7% lower risk of VTE, respectively. In addition, the correlation between sleep pattern and the incidence of VTE was consistent, regardless of genetic susceptibility (P for interaction = 0.366). CONCLUSIONS Our secondary analysis of a large-scale prospectively gathered registry revealed that individuals with a healthy sleep pattern are significantly correlated with lower risk of developing VTE, irrespective of genetic susceptibility.
Collapse
Affiliation(s)
- Jiaxin Bai
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ziyu Yang
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Jia
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Yu
- Department of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenli Jiang
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fanghui Li
- Department of Cardiology, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Rui Zeng
- Department of Cardiology, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Zhi Wan
- Department of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Lei
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyang Liao
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dongze Li
- Department of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qian Zhao
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Jee YH, Thibord F, Dominguez A, Sept C, Boulier K, Venkateswaran V, Ding Y, Cherlin T, Verma SS, Faro VL, Bartz TM, Boland A, Brody JA, Deleuze JF, Emmerich J, Germain M, Johnson AD, Kooperberg C, Morange PE, Pankratz N, Psaty BM, Reiner AP, Smadja DM, Sitlani CM, Suchon P, Tang W, Trégouët DA, Zöllner S, Pasaniuc B, Damrauer SM, Sanna S, Snieder H, Kabrhel C, Smith NL, Kraft P. Multi-ancestry polygenic risk scores for venous thromboembolism. Hum Mol Genet 2024; 33:1584-1591. [PMID: 38879759 PMCID: PMC11373328 DOI: 10.1093/hmg/ddae097] [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: 12/24/2023] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/25/2024] Open
Abstract
Venous thromboembolism (VTE) is a significant contributor to morbidity and mortality, with large disparities in incidence rates between Black and White Americans. Polygenic risk scores (PRSs) limited to variants discovered in genome-wide association studies in European-ancestry samples can identify European-ancestry individuals at high risk of VTE. However, there is limited evidence on whether high-dimensional PRS constructed using more sophisticated methods and more diverse training data can enhance the predictive ability and their utility across diverse populations. We developed PRSs for VTE using summary statistics from the International Network against Venous Thrombosis (INVENT) consortium genome-wide association studies meta-analyses of European- (71 771 cases and 1 059 740 controls) and African-ancestry samples (7482 cases and 129 975 controls). We used LDpred2 and PRS-CSx to construct ancestry-specific and multi-ancestry PRSs and evaluated their performance in an independent European- (6781 cases and 103 016 controls) and African-ancestry sample (1385 cases and 12 569 controls). Multi-ancestry PRSs with weights tuned in European-ancestry samples slightly outperformed ancestry-specific PRSs in European-ancestry test samples (e.g. the area under the receiver operating curve [AUC] was 0.609 for PRS-CSx_combinedEUR and 0.608 for PRS-CSxEUR [P = 0.00029]). Multi-ancestry PRSs with weights tuned in African-ancestry samples also outperformed ancestry-specific PRSs in African-ancestry test samples (PRS-CSxAFR: AUC = 0.58, PRS-CSx_combined AFR: AUC = 0.59), although this difference was not statistically significant (P = 0.34). The highest fifth percentile of the best-performing PRS was associated with 1.9-fold and 1.68-fold increased risk for VTE among European- and African-ancestry subjects, respectively, relative to those in the middle stratum. These findings suggest that the multi-ancestry PRS might be used to improve performance across diverse populations to identify individuals at highest risk for VTE.
Collapse
Affiliation(s)
- Yon Ho Jee
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, United States
| | - Florian Thibord
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, 31 Center Drive, Bethesda, MD 20892, United States
- Framingham Heart Study, Boston University and National Heart, Lung, and Blood Institute, Framingham, 73 Mt. Wayte Ave, Suite #2, Framingham, MA 01702, United States
| | - Alicia Dominguez
- Department of Biostatistics, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109, United States
| | - Corriene Sept
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, United States
| | - Kristin Boulier
- Bioinformatics Interdepartmental Program, University of California Los Angeles, 611 Charles E. Young Drive East, Los Angeles, CA 90095-1570, United States
| | - Vidhya Venkateswaran
- Department of Oral Biology, University of California Los Angeles School of Dentistry, 13-089 CHS, Box 951668, Box 951570, Los Angeles, CA 90095-1668, United States
| | - Yi Ding
- Bioinformatics Interdepartmental Program, University of California Los Angeles, 611 Charles E. Young Drive East, Los Angeles, CA 90095-1570, United States
| | - Tess Cherlin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, 3400 Spruce St. Philadelphia, PA 19104-4238, United States
| | - Shefali Setia Verma
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, 3400 Spruce St. Philadelphia, PA 19104-4238, United States
| | - Valeria Lo Faro
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Dag Hammarskjölds väg 20751 85 Uppsala, Sweden
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Departments of Biostatistics and Medicine, University of Washington, 4333 Brooklyn Ave, Seattle, WA 98195, United States
| | - Anne Boland
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, 91057 Evry, France
- Laboratory of Excellence in Medical Genomics, GENMED, F-91057 Evry, France
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, 4333 Brooklyn Ave, Seattle, WA 98195, United States
| | - Jean-Francois Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, 91057 Evry, France
- Laboratory of Excellence in Medical Genomics, GENMED, F-91057 Evry, France
- Centre d’Etude du Polymorphisme Humain, Fondation Jean Dausset, 27 rue Juliette Dodu, 75010 Paris, France
| | - Joseph Emmerich
- Department of Vascular Medicine, Paris Saint-Joseph Hospital Group, University of Paris, 75014 Paris, France
- INSERM CRESS UMR 1153, F-75005, Paris, France
| | - Marine Germain
- Bordeaux Population Health Research Center, University of Bordeaux, INSERM, UMR 1219, Bordeaux, France
| | - Andrew D Johnson
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, 31 Center Drive, Bethesda, MD 20892, United States
- Framingham Heart Study, Boston University and National Heart, Lung, and Blood Institute, Framingham, 73 Mt. Wayte Ave, Suite #2, Framingham, MA 01702, United States
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinbson Cancer Center, PO Box 19024, Seattle, WA 98109, United States
| | - Pierre-Emmanuel Morange
- Aix-Marseille University, INSERM, INRAE, Centre de Recherche en CardioVasculaire et Nutrition, Laboratory of Haematology, CRB Assistance Publique – Hôpitaux de Marseille, HemoVasc, 27, boulevard Jean Moulin, 13005 Marseille, France
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455, United States
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, 4333 Brooklyn Ave, Seattle, WA 98195, United States
- Department of Epidemiology, University of Washington, 4333 Brooklyn Ave, Seattle, WA 98195, United States
- Department of Health Systems and Population Health, University of Washington, 4333 Brooklyn Ave, Seattle, WA 98195, United States
| | - Alexander P Reiner
- Division of Public Health Sciences, Fred Hutchinbson Cancer Center, PO Box 19024, Seattle, WA 98109, United States
- Department of Epidemiology, University of Washington, 4333 Brooklyn Ave, Seattle, WA 98195, United States
| | - David M Smadja
- Innovative Therapies in Hemostasis, Université de Paris, INSERM, F-75006, Paris, France
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), F-75015, Paris, France
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, 4333 Brooklyn Ave, Seattle, WA 98195, United States
| | - Pierre Suchon
- Aix-Marseille University, INSERM, INRAE, Centre de Recherche en CardioVasculaire et Nutrition, Laboratory of Haematology, CRB Assistance Publique – Hôpitaux de Marseille, HemoVasc, 27, boulevard Jean Moulin, 13005 Marseille, France
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, 1300 S. 2nd St., Minneapolis, MN 55454, United States
| | - David-Alexandre Trégouët
- Bordeaux Population Health Research Center, University of Bordeaux, INSERM, UMR 1219, Bordeaux, France
| | - Sebastian Zöllner
- Department of Biostatistics, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109, United States
| | - Bogdan Pasaniuc
- Department of Oral Biology, University of California Los Angeles School of Dentistry, 13-089 CHS, Box 951668, Box 951570, Los Angeles, CA 90095-1668, United States
| | - Scott M Damrauer
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, 415 Curie Blvd, Philadelphia, PA 19104, United States
- Department of Surgery, Department of Genetics, and Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Building 421, Philadelphia, PA 19104, United States
- Department of Surgery, Corporal Michael Crescenz VA Medical Center, 3900 Woodland Ave, Philadelphia, PA 19104, United States
| | - Serena Sanna
- Department of Genetics, University of Groningen, University Medical Center Groningen (UMCG), PO Box 30.001, 9700 RB Groningen, The Netherlands
- Institute for Genetics and Biomedical Research, National Research Council, SS 554 Km 4,500, 09042 Monserrato CA, Italy
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Christopher Kabrhel
- Center for Vascular Emergencies, Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, United States
| | - Nicholas L Smith
- Department of Health Systems and Population Health, University of Washington, 4333 Brooklyn Ave, Seattle, WA 98195, United States
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, 1730 Minor Ave, Seattle, WA 98101, United States
- Department of Veterans Affairs Office of Research and Development, Seattle Epidemiologic Research and Information Center, 1660 S Columbian Way, S-152-E, Seattle, WA 98108, United States
| | - Peter Kraft
- Transdivisional Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr, Rockville, MD 20850, United States
| | | |
Collapse
|
19
|
Underwood M, Bidlack C, Desch KC. Venous thromboembolic disease genetics: from variants to function. J Thromb Haemost 2024; 22:2393-2403. [PMID: 38908832 PMCID: PMC11934295 DOI: 10.1016/j.jtha.2024.06.004] [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: 05/03/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/24/2024]
Abstract
Venous thromboembolic disease (VTE) is a prevalent and potentially life-threatening vascular disease, including both deep vein thrombosis and pulmonary embolism. This review will focus on recent insights into the heritable factors that influence an individual's risk for VTE. Here, we will explore not only the discovery of new genetic risk variants but also the importance of functional characterization of these variants. These genome-wide studies should lead to a better understanding of the biological role of genes inside and outside of the canonical coagulation system in thrombus formation and lead to an improved ability to predict an individual's risk of VTE. Further understanding of the molecular mechanisms altered by genetic variation in VTE risk will be accelerated by further human genome sequencing efforts and the use of functional genetic screens.
Collapse
Affiliation(s)
- Mary Underwood
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher Bidlack
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Karl C Desch
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA; Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
20
|
Athar A, Kashyap P, Khan S, Sattar RSA, Khan SA, Prasad S, Husain SA, Parveen F. Genetic landscape of thrombophilia in recurrent miscarriages. Obstet Gynecol Sci 2024; 67:435-448. [PMID: 39069307 PMCID: PMC11424186 DOI: 10.5468/ogs.22084] [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: 03/12/2022] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
The etiology of recurrent miscarriage (RM) is extremely heterogeneous, encompassing genetic, immunological, anatomical, endocrine, thrombophilic, infectious, and uterine abnormalities. Thrombophilia is a major contributor to pregnancy complications, potentially harming the fetus and jeopardizing the continuation of pregnancy. Therefore, successful pregnancy outcomes depend on maintaining a delicate balance between coagulation and fibrinolytic factors, crucial for ensuring the adjustment of the basal plate to facilitate adequate placental perfusion. Despite numerous studies shedding light on the role of thrombophilic factors and genetic variations in RM, the exact pathogenesis remains unclear. It is imperative to systematically rule out thrombophilia and other related factors responsible for pregnancy disorders and RMs to guide appropriate and active management strategies. Addressing thrombophilia continues to present challenges in terms of effective treatment. The current review aims to address the heterogeneity of RM as a therapeutic challenge, emphasizing the need for standardized diagnostic tests and welldesigned multicenter research trials to gather robust, evidence-based data on thrombophilic causes of RM and provide effective treatment. The goal is to enhance the understanding of thrombophilic factors and genetic landscapes associated with RM through various approaches, including candidate gene studies, genome-wide association studies, and high-throughput sequencing. Meta-analyses have underscored the significance of genetic aberrations in RM, highlighting the necessity for identifying critical mutations implicated in the etiopathogenesis of miscarriages to pave the way for implementation of targeted clinical therapies.
Collapse
Affiliation(s)
- Alina Athar
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Poonam Kashyap
- Department of Obstetrics and Gynaecology, Maulana Azad Medical College, New Delhi, India
| | - Shagufta Khan
- Department of Pathology, Era's Medical College, Lucknow, India
| | | | | | - Sudha Prasad
- Department of Pathology, Era's Medical College, Lucknow, India
| | | | - Farah Parveen
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
21
|
Hasser EK, Brody JA, Bartz TM, Thibord F, Li-Gao R, Kauko A, Wiggins KL, Teder-Laving M, Kim J, Munsch G, Haile HG, Deleuze JF, van Hylckama Vlieg A, Wolberg AS, Boland A, Morange PE, Kraft P, Lowenstein CJ, Emmerich J, Sitlani CM, Suchon P, Rosendaal FR, Niiranen T, Kabrhel C, Trégouët DA, Smith NL. Genome-wide investigation of exogenous female hormones, genetic variation, and venous thromboembolism risk. J Thromb Haemost 2024; 22:2261-2269. [PMID: 38782299 DOI: 10.1016/j.jtha.2024.05.011] [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: 12/12/2023] [Revised: 04/02/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Increased risk of venous thromboembolism (VTE) is a life-threatening side effect for users of oral contraceptives (OCs) or hormone therapy (HT). OBJECTIVES To investigate the potential for genetic predisposition to VTE in OC or HT users, we conducted a gene-by-environment case-only meta-analysis of genome-wide association studies (GWAS). METHODS Use or nonuse of OCs (7 studies) or HT (8 studies) at the time of the VTE event was determined by pharmacy records or self-report. A synergy index (SI) was modeled for each variant in each study and submultiplicative/supramultiplicative gene-by-environment interactions were estimated. The SI parameters were first meta-analyzed across OC and HT studies and subsequently meta-analyzed to obtain an overall estimate. The primary analysis was agnostic GWAS and interrogated all imputed genotypes using a P value threshold of <5.0 × 10-8; secondary analyses were candidate-based. RESULTS The VTE case-only OC meta-analysis included 2895 OC users and 6607 nonusers; the case-only HT meta-analysis included 2434 HT users and 12 793 nonusers. In primary GWAS meta-analyses, no variant reached genome-wide significance, but the smallest P value approached statistical significance: rs9386463 (P = 5.03 × 10-8). We tested associations for 138 candidate variants and identified 2 that exceeded statistical significance (0.05/138 = 3.62 × 10-4): F5 rs6025 (P = 1.87 × 10-5; SI, 1.29; previously observed) and F11 rs2036914 (P = 2.0 × 10-4; SI, 0.91; new observation). CONCLUSION The candidate variant approach to identify submultiplictive/supramultiplicative associations between genetic variation and OC and HT use identified a new association with common genetic variation in F11, while the agnostic interrogations did not yield new discoveries.
Collapse
Affiliation(s)
- Emily K Hasser
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Traci M Bartz
- Departments of Biostatistics and Medicine, University of Washington, Seattle, WA, USA
| | - Florian Thibord
- INSERM, Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Bordeaux, France; Laboratory of Excellence on Medical Genomics (GENMED), Evry, France
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anni Kauko
- Department of Internal Medicine, University of Turku, Turku, Finland
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Jihye Kim
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gaëlle Munsch
- INSERM, Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Bordeaux, France
| | - Helen G Haile
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Jean-Francois Deleuze
- CEA, Centre National de Recherche en Génomique Humaine (CNRGH), Université Paris-Saclay, Evry, France; Centre D'Etude du Polymorphisme Humain, Fondation Jean Dausset, Paris, France; Laboratory of Excellence on Medical Genomics (GENMED), Evry, France
| | | | - Alisa S Wolberg
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anne Boland
- CEA, Centre National de Recherche en Génomique Humaine (CNRGH), Université Paris-Saclay, Evry, France; Laboratory of Excellence on Medical Genomics (GENMED), Evry, France
| | - Pierre-Emmanuel Morange
- Hematology Laboratory, La Timone University Hospital of Marseille, Marseille, France; Centre de recherche en CardioVasculaire et Nutrition, INSERM, INRAE, Aix-Marseille University, Marseille, France
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Joseph Emmerich
- Department of Vascular Medicine, Saint-Joseph Hospital Group, University of Paris Cité, Paris, France; UMR1153, INSERM Centre for Research in Epidemiology and Statistics (CRESS), Paris, France
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Pierre Suchon
- Hematology Laboratory, La Timone University Hospital of Marseille, Marseille, France; Centre de recherche en CardioVasculaire et Nutrition, INSERM, INRAE, Aix-Marseille University, Marseille, France
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Teemu Niiranen
- Department of Internal Medicine, University of Turku, Turku, Finland; Department of Internal Medicine, Division of Medicine, Turku University Hospital, Turku, Finland; Department of Public Health Solutions, Finish Institute of Health and Welfare, Helsinki, Finland
| | - Christopher Kabrhel
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Emergency Medicine, Harvard Medical School, Boston, MA, USA
| | - David-Alexandre Trégouët
- INSERM, Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, Bordeaux, France; Laboratory of Excellence on Medical Genomics (GENMED), Evry, France
| | - Nicholas L Smith
- Department of Epidemiology, University of Washington, Seattle, WA, USA; Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, USA; Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle, WA, USA.
| |
Collapse
|
22
|
Tan MCB, Isom CA, Liu Y, Trégouët DA, Wu L, Zhou D, Gamazon ER. Transcriptome-wide association study and Mendelian randomization in pancreatic cancer identifies susceptibility genes and causal relationships with type 2 diabetes and venous thromboembolism. EBioMedicine 2024; 106:105233. [PMID: 39002386 PMCID: PMC11284564 DOI: 10.1016/j.ebiom.2024.105233] [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: 01/18/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 07/15/2024] Open
Abstract
BACKGROUND Two important questions regarding the genetics of pancreatic adenocarcinoma (PDAC) are 1. Which germline genetic variants influence the incidence of this cancer; and 2. Whether PDAC causally predisposes to associated non-malignant phenotypes, such as type 2 diabetes (T2D) and venous thromboembolism (VTE). METHODS In this study of 8803 patients with PDAC and 67,523 controls, we first performed a large-scale transcriptome-wide association study to investigate the association between genetically determined gene expression in normal pancreas tissue and PDAC risk. Secondly, we used Mendelian Randomization (MR) to analyse the causal relationships among PDAC, T2D (74,124 cases and 824,006 controls) and VTE (30,234 cases and 172,122 controls). FINDINGS Sixteen genes showed an association with PDAC risk (FDR <0.10), including six genes not yet reported for PDAC risk (PPIP5K2, TFR2, HNF4G, LRRC10B, PRC1 and FBXL20) and ten previously reported genes (INHBA, SMC2, ABO, PDX1, MTMR6, ACOT2, PGAP3, STARD3, GSDMB, ADAM33). MR provided support for a causal effect of PDAC on T2D using genetic instruments in the HNF4G and PDX1 loci, and unidirectional causality of VTE on PDAC involving the ABO locus (OR 2.12, P < 1e-7). No evidence of a causal effect of PDAC on VTE was found. INTERPRETATION These analyses identified candidate susceptibility genes and disease relationships for PDAC that warrant further investigation. HNF4G and PDX1 may induce PDAC-associated diabetes, whereas ABO may induce the causative effect of VTE on PDAC. FUNDING National Institutes of Health (USA).
Collapse
Affiliation(s)
- Marcus C B Tan
- Division of Surgical Oncology and Endocrine Surgery, Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Chelsea A Isom
- Herbert Wertheim School of Public Health & Human Longevity Science, University of California, San Diego, San Diego, CA, USA
| | - Yangzi Liu
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Lang Wu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Dan Zhou
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China.
| | - Eric R Gamazon
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Clare Hall, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
23
|
Sundler Björkman L, Pirouzifard M, Grover SP, Egesten A, Sundquist J, Sundquist K, Zöller B. Increased risk of venous thromboembolism in young and middle-aged individuals with hereditary angioedema: a family study. Blood 2024; 144:435-444. [PMID: 38767511 DOI: 10.1182/blood.2023022996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/23/2024] [Accepted: 04/10/2024] [Indexed: 05/22/2024] Open
Abstract
ABSTRACT Hereditary angioedema (HAE), caused by C1 inhibitor protein deficiency, was recently shown to be associated with an increased risk for venous thromboembolism (VTE). To our knowledge, this is the first national family study of HAE, which aimed to determine the familial risk of VTE. The Swedish Multi-Generation Register was linked to the Swedish National Patient Register for the period of 1964 to 2018. Only patients with HAE with a validated diagnosis were included in the study and were linked to their family members. Hazard ratios (HRs) and 95% confidence intervals (CIs) for VTE were calculated for patients with HAE in comparison with relatives without HAE. Among 2006 individuals (from 276 pedigrees of 365 patients with HAE), 103 individuals were affected by VTE. In total, 35 (9.6%) patients with HAE were affected by VTE, whereas 68 (4.1%) non-HAE relatives were affected (P < .001). The adjusted HR for VTE among patients with HAE was 2.51 (95% CI, 1.67-3.77). Patients with HAE were younger at the first VTE than their non-HAE relatives (mean age, 51 years vs 63 years; P < .001). Before the age of 70 years, the HR for VTE among patients with HAE was 3.62 (95% CI, 2.26-5.80). The HR for VTE for patients with HAE born after 1964 was 8.29 (95% CI, 2.90-23.71). The HR for VTE for patients with HAE who were born in 1964 or earlier was 1.82 (95% CI, 1.14-2.91). HAE is associated with VTE among young and middle-aged individuals in Swedish families with HAE. The effect size of the association is in the order of other thrombophilias. We suggest that HAE may be considered a new rare thrombophilia.
Collapse
Affiliation(s)
- Linda Sundler Björkman
- Respiratory Medicine, Allergology, and Palliative Medicine, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
- Division of Hematology, Department of Medicine, UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - MirNabi Pirouzifard
- Department of Clinical Sciences Malmö, Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden
| | - Steven P Grover
- Division of Hematology, Department of Medicine, UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Arne Egesten
- Respiratory Medicine, Allergology, and Palliative Medicine, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Jan Sundquist
- Department of Clinical Sciences Malmö, Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden
| | - Kristina Sundquist
- Department of Clinical Sciences Malmö, Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden
| | - Bengt Zöller
- Department of Clinical Sciences Malmö, Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden
| |
Collapse
|
24
|
Sun Y, Deng J, Ding Y, Luo S, Li S, Guan Y, Cao X, Hao X, Hu Y. Serum albumin, genetic susceptibility, and risk of venous thromboembolism. Res Pract Thromb Haemost 2024; 8:102509. [PMID: 39221449 PMCID: PMC11364004 DOI: 10.1016/j.rpth.2024.102509] [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: 03/13/2024] [Accepted: 07/03/2024] [Indexed: 09/04/2024] Open
Abstract
Background Previous research on the association between serum albumin (ALB) and venous thromboembolism (VTE) has produced inconclusive results. The polygenic risk score is constructed from a set of independent risk variants associated with a disorder, enabling the identification of a larger fraction of the population at comparable or greater disease risk. It is still unknown whether ALB and genetic factors jointly contribute to the incidence of VTE. Objectives The present study aimed to explore ALB, genetic susceptibility, and the risk of VTE. Methods The present investigation was an analysis of prospectively collected data from UK Biobank, a population-based, longitudinal cohort. Cox proportional models were used to calculate hazard ratios and 95% CIs for VTE. The Kaplan-Meier curve was utilized to visualize the cumulative risk of VTE according to different serum ALB levels, and the restricted cubic spline model was leveraged to explore the exposure-response relationship among ALB levels and VTE risk. Results During median follow-up of 13.5 years, 11,502 cases with VTE were diagnosed among 417,113 participants in the UK Biobank. The lower ALB levels were associated with a higher risk for VTE. Individuals with both a high genetic risk and lowest ALB level had the highest risk of VTE (hazard ratio, 3.89; 95% CI, 3.41-4.43), compared with those with low genetic risk and highest ALB level. The positive joint effects of low ALB and polygenic risk score increased the risk of VTE in individuals with high genetic risk. This study excluded non-European patients and primarily focused on the European population, which may limit the generalizability of the findings. Conclusion Low serum ALB levels were linked to an increased risk of VTE, which was in accordance with a linear dose-response relationship. There was a positive additive effect of ALB and genetic susceptibility on the risk of VTE. ALB could serve as a biomarker for predicting the risk of VTE.
Collapse
Affiliation(s)
- Yuyang Sun
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Deng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yajie Ding
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shanshan Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Si Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yunlong Guan
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xi Cao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
25
|
Jiang C, Lin J, Xie B, Peng M, Dai Z, Mai S, Chen Q. Causal association between circulating blood cell traits and pulmonary embolism: a mendelian randomization study. Thromb J 2024; 22:49. [PMID: 38863024 PMCID: PMC11167760 DOI: 10.1186/s12959-024-00618-3] [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: 03/20/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Pulmonary embolism (PE) is a life-threatening thromboembolic disease for which there is limited evidence for effective prevention and treatment. Our goal was to determine whether genetically predicted circulating blood cell traits could influence the incidence of PE. METHODS Using single variable Mendelian randomization (SVMR) and multivariate Mendelian randomization (MVMR) analyses, we identified genetic associations between circulating blood cell counts and lymphocyte subsets and PE. GWAS blood cell characterization summary statistics were compiled from the Blood Cell Consortium. The lymphocyte subpopulation counts were extracted from summary GWAS statistics for samples from 3757 individuals that had been analyzed by flow cytometry. GWAS data related to PE were obtained from the FinnGen study. RESULTS According to the SVMR and reverse MR, increased levels of circulating white blood cells (odds ratio [OR]: 0.88, 95% confidence interval [CI]: 0.81-0.95, p = 0.0079), lymphocytes (OR: 0.90, 95% CI: 0.84-0.97, p = 0.0115), and neutrophils (OR: 0.88, 95% CI: 0.81-0.96, p = 0.0108) were causally associated with PE susceptibility. MVMR analysis revealed that lower circulating lymphocyte counts (OR: 0.84, 95% CI: 0.75-0.94, p = 0.0139) were an independent predictor of PE. According to further MR results, this association may be primarily related to HLA-DR+ natural killer (NK) cells. CONCLUSIONS Among European populations, there is a causal association between genetically predicted low circulating lymphocyte counts, particularly low HLA-DR+ NK cells, and an increased risk of PE. This finding supports observational studies that link peripheral blood cells to PE and provides recommendations for predicting and preventing this condition.
Collapse
Affiliation(s)
- Chen Jiang
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jianing Lin
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Bin Xie
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Meijuan Peng
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ziyu Dai
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Suyin Mai
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiong Chen
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
26
|
Ryu J, Rämö JT, Jurgens SJ, Niiranen T, Sanna-Cherchi S, Bauer KA, Haj A, Choi SH, Palotie A, Daly M, Ellinor PT, Bendapudi PK. Thrombosis risk in single- and double-heterozygous carriers of factor V Leiden and prothrombin G20210A in FinnGen and the UK Biobank. Blood 2024; 143:2425-2432. [PMID: 38498041 PMCID: PMC11830983 DOI: 10.1182/blood.2023023326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/19/2024] Open
Abstract
ABSTRACT The factor V Leiden (FVL; rs6025) and prothrombin G20210A (PTGM; rs1799963) polymorphisms are 2 of the most well-studied genetic risk factors for venous thromboembolism (VTE). However, double heterozygosity (DH) for FVL and PTGM remains poorly understood, with previous studies showing marked disagreement regarding thrombosis risk conferred by the DH genotype. Using multidimensional data from the UK Biobank (UKB) and FinnGen biorepositories, we evaluated the clinical impact of DH carrier status across 937 939 individuals. We found that 662 participants (0.07%) were DH carriers. After adjustment for age, sex, and ancestry, DH individuals experienced a markedly elevated risk of VTE compared with wild-type individuals (odds ratio [OR] = 5.24; 95% confidence interval [CI], 4.01-6.84; P = 4.8 × 10-34), which approximated the risk conferred by FVL homozygosity. A secondary analysis restricted to UKB participants (N = 445 144) found that effect size estimates for the DH genotype remained largely unchanged (OR = 4.53; 95% CI, 3.42-5.90; P < 1 × 10-16) after adjustment for commonly cited VTE risk factors, such as body mass index, blood type, and markers of inflammation. In contrast, the DH genotype was not associated with a significantly higher risk of any arterial thrombosis phenotype, including stroke, myocardial infarction, and peripheral artery disease. In summary, we leveraged population-scale genomic data sets to conduct, to our knowledge, the largest study to date on the DH genotype and were able to establish far more precise effect size estimates than previously possible. Our findings indicate that the DH genotype may occur as frequently as FVL homozygosity and may confer a similarly increased risk of VTE.
Collapse
Affiliation(s)
- Justine Ryu
- Department of Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Joel T. Rämö
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Cardiology Division, Massachusetts General Hospital, Boston, MA
| | - Sean J. Jurgens
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA
- Cardiology Division, Massachusetts General Hospital, Boston, MA
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - 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
| | | | - Kenneth A. Bauer
- Harvard Medical School, Boston, MA
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, MA
| | - Amelia Haj
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Seung Hoan Choi
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Aarno Palotie
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Cardiology Division, Massachusetts General Hospital, Boston, MA
| | - Mark Daly
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Cardiology Division, Massachusetts General Hospital, Boston, MA
| | - Patrick T. Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA
- Cardiology Division, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Pavan K. Bendapudi
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, MA
- Division of Hematology and Blood Transfusion Service, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
27
|
Han X, Knauss EA, Fuente MDL, Li W, Conlon RA, LePage DF, Jiang W, Renna SA, McKenzie SE, Nieman MT. A mouse model of the protease-activated receptor 4 Pro310Leu variant has reduced platelet reactivity. J Thromb Haemost 2024; 22:1715-1726. [PMID: 38508397 PMCID: PMC12036797 DOI: 10.1016/j.jtha.2024.03.004] [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: 12/01/2023] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Protease-activated receptor 4 (PAR4) mediates thrombin signaling on platelets and other cells. Our recent structural studies demonstrated that a single nucleotide polymorphism in extracellular loop 3 and PAR4-P310L (rs2227376) leads to a hyporeactive receptor. OBJECTIVES The goal of this study was to determine how the hyporeactive PAR4 variant in extracellular loop 3 impacts platelet function in vivo using a novel knock-in mouse model (PAR4-322L). METHODS A point mutation was introduced into the PAR4 gene F2rl3 via CRISPR/Cas9 to create PAR4-P322L, the mouse homolog to human PAR4-P310L. Platelet response to PAR4 activation peptide (AYPGKF), thrombin, ADP, and convulxin was monitored by αIIbβ3 integrin activation and P-selectin translocation using flow cytometry or platelet aggregation. In vivo responses were determined by the tail bleeding assay and the ferric chloride-induced carotid artery injury model. RESULTS PAR4-P/L and PAR4-L/L platelets had a reduced response to AYPGKF and thrombin measured by P-selectin translocation or αIIbβ3 activation. The response to ADP and convulxin was unchanged among genotypes. In addition, both PAR4-P/L and PAR4-L/L platelets showed a reduced response to thrombin in aggregation studies. There was an increase in the tail bleeding time for PAR4-L/L mice. The PAR4-P/L and PAR4-L/L mice both showed an extended time to arterial thrombosis. CONCLUSION PAR4-322L significantly reduced platelet responsiveness to AYPGKF and thrombin, which is in agreement with our previous structural and cell signaling studies. In addition, PAR4-322L had prolonged arterial thrombosis time. Our mouse model provides a foundation to further evaluate the role of PAR4 in other pathophysiological contexts.
Collapse
Affiliation(s)
- Xu Han
- Case Western Reserve University School of Medicine, Department of Pharmacology, Cleveland, Ohio, USA
| | - Elizabeth A Knauss
- Case Western Reserve University School of Medicine, Department of Pharmacology, Cleveland, Ohio, USA
| | - Maria de la Fuente
- Case Western Reserve University School of Medicine, Department of Pharmacology, Cleveland, Ohio, USA
| | - Wei Li
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, West Virginia, USA
| | - Ronald A Conlon
- Case Transgenic and Targeting Facility, Case Western Reserve University, Cleveland, Ohio, USA
| | - David F LePage
- Case Transgenic and Targeting Facility, Case Western Reserve University, Cleveland, Ohio, USA
| | - Weihong Jiang
- Case Transgenic and Targeting Facility, Case Western Reserve University, Cleveland, Ohio, USA
| | - Stephanie A Renna
- Department of Medicine, The Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Steven E McKenzie
- Department of Medicine, The Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Marvin T Nieman
- Case Western Reserve University School of Medicine, Department of Pharmacology, Cleveland, Ohio, USA.
| |
Collapse
|
28
|
Jiang Z, Sullivan PF, Li T, Zhao B, Wang X, Luo T, Huang S, Guan PY, Chen J, Yang Y, Stein JL, Li Y, Liu D, Sun L, Zhu H. The pivotal role of the X-chromosome in the genetic architecture of the human brain. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.08.30.23294848. [PMID: 37693466 PMCID: PMC10491353 DOI: 10.1101/2023.08.30.23294848] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Genes on the X-chromosome are extensively expressed in the human brain. However, little is known for the X-chromosome's impact on the brain anatomy, microstructure, and functional network. We examined 1,045 complex brain imaging traits from 38,529 participants in the UK Biobank. We unveiled potential autosome-X-chromosome interactions, while proposing an atlas outlining dosage compensation (DC) for brain imaging traits. Through extensive association studies, we identified 72 genome-wide significant trait-locus pairs (including 29 new associations) that share genetic architectures with brain-related disorders, notably schizophrenia. Furthermore, we discovered unique sex-specific associations and assessed variations in genetic effects between sexes. Our research offers critical insights into the X-chromosome's role in the human brain, underscoring its contribution to the differences observed in brain structure and functionality between sexes.
Collapse
|
29
|
Turizo MJF, Patell R, Zwicker JI. Identifying novel biomarkers using proteomics to predict cancer-associated thrombosis. BLEEDING, THROMBOSIS AND VASCULAR BIOLOGY 2024; 3:120. [PMID: 38828226 PMCID: PMC11143428 DOI: 10.4081/btvb.2024.120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/08/2024] [Indexed: 06/05/2024]
Abstract
Comprehensive protein analyses of plasma are made possible by high-throughput proteomic screens, which may help find new therapeutic targets and diagnostic biomarkers. Patients with cancer are frequently affected by venous thromboembolism (VTE). The limited predictive accuracy of current VTE risk assessment tools highlights the need for new, more targeted biomarkers. Although coagulation biomarkers for the diagnosis, prognosis, and treatment of VTE have been investigated, none of them have the necessary clinical validation or diagnostic accuracy. Proteomics holds the potential to uncover new biomarkers and thrombotic pathways that impact the risk of thrombosis. This review explores the fundamental methods used in proteomics and focuses on particular biomarkers found in VTE and cancer-associated thrombosis.
Collapse
Affiliation(s)
- Maria J Fernandez Turizo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Rushad Patell
- Division of Medical Oncology and Hematology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Jeffrey I Zwicker
- Department of Medicine, Hematology Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Weil Cornell Medical College, New York, NY, United States
| |
Collapse
|
30
|
Ghorbani A, Greathouse J, Bakhshaei S, Ghorbani A, Zamiri K, Ho L, Ho A. Persistent Peril: Recurrent Deep Vein Thrombosis and Pulmonary Embolism in a Patient With Protein S Deficiency Despite Optimal Anticoagulation Therapies. Cureus 2024; 16:e60517. [PMID: 38883011 PMCID: PMC11180491 DOI: 10.7759/cureus.60517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
The clotting system has evolved as an adaptive mechanism to prevent blood loss during vascular damage. However, the intricate nature of the clotting cascade and the complexities of human life can sometimes lead to the unnatural activation of this delicate cascade. This can result in blood clot formation within the cardiovascular system, contributing to a wide range of pathological conditions. Abnormal intravascular coagulation most commonly occurs in the deep veins of the lower extremities, and can emboli to other organs, hence, it is termed "venous thromboembolism" (VTE). In this report, we introduce a challenging case of VTE that poses a dilemma for current medical management. The patient with possible protein S deficiency underwent various guideline-directed medical treatments, yet experienced recurrent VTE episodes, including deep vein thrombosis (DVT) and pulmonary embolism (PE), leading to hospital readmissions. This case report sheds light on our challenges in effectively treating VTE.
Collapse
Affiliation(s)
- Ali Ghorbani
- Internal Medicine, Southwest Healthcare, Temecula, USA
| | | | - Sina Bakhshaei
- Internal Medicine, Southern California Medical Education Consortium, Temecula Valley Hospital, Temecula, USA
| | - Aida Ghorbani
- Neurology, UCLA School of Medicine, Los Angeles, USA
| | - Kurosh Zamiri
- Biology, University of California Los Angeles, Los Angeles, USA
| | - Lauren Ho
- Cardiology, Temecula Valley Hospital, Temecula, USA
| | - Andrew Ho
- Cardiology, Temecula Valley Hospital, Temecula, USA
| |
Collapse
|
31
|
Mathey CM, Maj C, Eriksson N, Krebs K, Westmeier J, David FS, Koromina M, Scheer AB, Szabo N, Wedi B, Wieczorek D, Amann PM, Löffler H, Koch L, Schöffl C, Dickel H, Ganjuur N, Hornung T, Buhl T, Greve J, Wurpts G, Aygören-Pürsün E, Steffens M, Herms S, Heilmann-Heimbach S, Hoffmann P, Schmidt B, Mavarani L, Andresen T, Sørensen SB, Andersen V, Vogel U, Landén M, Bulik CM, Bygum A, Magnusson PKE, von Buchwald C, Hallberg P, Rye Ostrowski S, Sørensen E, Pedersen OB, Ullum H, Erikstrup C, Bundgaard H, Milani L, Rasmussen ER, Wadelius M, Ghouse J, Sachs B, Nöthen MM, Forstner AJ. Meta-analysis of ACE inhibitor-induced angioedema identifies novel risk locus. J Allergy Clin Immunol 2024; 153:1073-1082. [PMID: 38300190 DOI: 10.1016/j.jaci.2023.11.921] [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: 05/31/2023] [Revised: 10/20/2023] [Accepted: 11/13/2023] [Indexed: 02/02/2024]
Abstract
BACKGROUND Angioedema is a rare but potentially life-threatening adverse drug reaction in patients receiving angiotensin-converting enzyme inhibitors (ACEis). Research suggests that susceptibility to ACEi-induced angioedema (ACEi-AE) involves both genetic and nongenetic risk factors. Genome- and exome-wide studies of ACEi-AE have identified the first genetic risk loci. However, understanding of the underlying pathophysiology remains limited. OBJECTIVE We sought to identify further genetic factors of ACEi-AE to eventually gain a deeper understanding of its pathophysiology. METHODS By combining data from 8 cohorts, a genome-wide association study meta-analysis was performed in more than 1000 European patients with ACEi-AE. Secondary bioinformatic analyses were conducted to fine-map associated loci, identify relevant genes and pathways, and assess the genetic overlap between ACEi-AE and other traits. Finally, an exploratory cross-ancestry analysis was performed to assess shared genetic factors in European and African-American patients with ACEi-AE. RESULTS Three genome-wide significant risk loci were identified. One of these, located on chromosome 20q11.22, has not been implicated previously in ACEi-AE. Integrative secondary analyses highlighted previously reported genes (BDKRB2 [bradykinin receptor B2] and F5 [coagulation factor 5]) as well as biologically plausible novel candidate genes (PROCR [protein C receptor] and EDEM2 [endoplasmic reticulum degradation enhancing alpha-mannosidase like protein 2]). Lead variants at the risk loci were found with similar effect sizes and directions in an African-American cohort. CONCLUSIONS The present results contributed to a deeper understanding of the pathophysiology of ACEi-AE by (1) providing further evidence for the involvement of bradykinin signaling and coagulation pathways and (2) suggesting, for the first time, the involvement of the fibrinolysis pathway in this adverse drug reaction. An exploratory cross-ancestry comparison implicated the relevance of the associated risk loci across diverse ancestries.
Collapse
Affiliation(s)
- Carina M Mathey
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Carlo Maj
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Bonn, Germany; Centre for Human Genetics, University of Marburg, Marburg, Germany
| | - Niclas Eriksson
- Uppsala Clinical Research Center, Uppsala, Sweden; Department of Medical Sciences, Clinical Pharmacogenomics and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Kristi Krebs
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Julia Westmeier
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Friederike S David
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | | | - Annika B Scheer
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Nora Szabo
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Bettina Wedi
- Department of Dermatology and Allergy, Comprehensive Allergy Center, Hannover Medical School, Hannover, Germany
| | - Dorothea Wieczorek
- Department of Dermatology and Allergy, Comprehensive Allergy Center, Hannover Medical School, Hannover, Germany
| | - Philipp M Amann
- Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, Krems, Austria
| | - Harald Löffler
- Department of Dermatology, SLK Hospital Heilbronn, Heilbronn, Germany
| | - Lukas Koch
- Department of Dermatology and Venereology, Medical University Graz, Graz, Austria
| | - Clemens Schöffl
- Department of Dermatology and Venereology, Medical University Graz, Graz, Austria
| | - Heinrich Dickel
- Department of Dermatology, Venereology and Allergology, St Josef Hospital, University Medical Center, Ruhr University Bochum, Bochum, Germany
| | - Nomun Ganjuur
- Department of Dermatology, Venereology and Allergology, St Josef Hospital, University Medical Center, Ruhr University Bochum, Bochum, Germany; Institute of Health Care Research in Dermatology and Nursing (IVDP), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Hornung
- Department of Dermatology and Allergy, University Hospital of Bonn, Bonn, Germany
| | - Timo Buhl
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| | - Jens Greve
- Department of Otorhinolaryngology-Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Gerda Wurpts
- Department of Dermatology and Allergy, Aachen Comprehensive Allergy Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Emel Aygören-Pürsün
- Department for Children and Adolescents, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Michael Steffens
- Research Division, Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Stefan Herms
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Stefanie Heilmann-Heimbach
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Börge Schmidt
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Laven Mavarani
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Trine Andresen
- Molecular Diagnostics and Clinical Research Unit, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Signe Bek Sørensen
- Molecular Diagnostics and Clinical Research Unit, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Vibeke Andersen
- Molecular Diagnostics and Clinical Research Unit, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark; Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark; OPEN, University of Southern Denmark, Odense, Denmark
| | - Ulla Vogel
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Mikael Landén
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Cynthia M Bulik
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Anette Bygum
- Department of Clinical Institute, University of Southern Denmark, Odense, Denmark; Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Christian von Buchwald
- Department of Otorhinolaryngology-Head and Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Pär Hallberg
- Department of Medical Sciences, Clinical Pharmacogenomics and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen Hospital Biobank Unit, Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen Hospital Biobank Unit, Rigshospitalet, Copenhagen, Denmark
| | - Ole B Pedersen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Christian Erikstrup
- Departments of Clinical Immunology, Aarhus University, Aarhus, Denmark; Departments of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Henning Bundgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lili Milani
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Eva Rye Rasmussen
- Department of Otorhinolaryngology-Head and Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Departments of Private Practice Ølsemaglevej, Køge, Denmark
| | - Mia Wadelius
- Department of Medical Sciences, Clinical Pharmacogenomics and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jonas Ghouse
- Laboratory for Molecular Cardiology, Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Laboratory for Molecular Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bernhardt Sachs
- Department of Dermatology and Allergy, Aachen Comprehensive Allergy Center, University Hospital RWTH Aachen, Aachen, Germany; Research Division, Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Andreas J Forstner
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany; Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, Germany.
| |
Collapse
|
32
|
He XY, Wu BS, Yang L, Guo Y, Deng YT, Li ZY, Fei CJ, Liu WS, Ge YJ, Kang J, Feng J, Cheng W, Dong Q, Yu JT. Genetic associations of protein-coding variants in venous thromboembolism. Nat Commun 2024; 15:2819. [PMID: 38561338 PMCID: PMC10984941 DOI: 10.1038/s41467-024-47178-8] [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: 07/12/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
Previous genetic studies of venous thromboembolism (VTE) have been largely limited to common variants, leaving the genetic determinants relatively incomplete. We performed an exome-wide association study of VTE among 14,723 cases and 334,315 controls. Fourteen known and four novel genes (SRSF6, PHPT1, CGN, and MAP3K2) were identified through protein-coding variants, with broad replication in the FinnGen cohort. Most genes we discovered exhibited the potential to predict future VTE events in longitudinal analysis. Notably, we provide evidence for the additive contribution of rare coding variants to known genome-wide polygenic risk in shaping VTE risk. The identified genes were enriched in pathways affecting coagulation and platelet activation, along with liver-specific expression. The pleiotropic effects of these genes indicated the potential involvement of coagulation factors, blood cell traits, liver function, and immunometabolic processes in VTE pathogenesis. In conclusion, our study unveils the valuable contribution of protein-coding variants in VTE etiology and sheds new light on its risk stratification.
Collapse
Affiliation(s)
- Xiao-Yu He
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yue-Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ze-Yu Li
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Chen-Jie Fei
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei-Shi Liu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Jun Ge
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jujiao Kang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
- Department of Computer Science, University of Warwick, Coventry, UK
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
- Department of Computer Science, University of Warwick, Coventry, UK.
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
33
|
Zöller B, Sundquist J, Sundquist K, Ohlsson H. The risk for venous thromboembolism and cardiometabolic disorders in offspring from thrombosis-prone pedigrees. J Thromb Haemost 2024; 22:775-784. [PMID: 38072377 DOI: 10.1016/j.jtha.2023.11.024] [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: 08/12/2023] [Revised: 11/04/2023] [Accepted: 11/27/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Most family studies on venous thromboembolism (VTE) have focused on first-degree relatives. OBJECTIVES We took a pedigree-based approach and examined the risk of VTE and cardiometabolic disorders in offspring from extended pedigrees according to the densities of VTE in pedigrees. METHODS From the Swedish population, we identified a total of 482 185 pedigrees containing a mean of 14.2 parents, aunts/uncles, grandparents, and cousins of a core full sibship that we termed the pedigree offspring (n = 751 060). We then derived 8 empirical classes of these pedigrees based on the density of cases of VTE. The risk was determined in offspring for VTE and cardiometabolic disorders as a function of VTE density in their pedigrees. Bonferroni correction for multiple comparisons was performed. RESULTS VTE was unevenly distributed in the population; the Gini coefficient was 0.59. Higher VTE density in pedigrees was associated in the offspring with a higher risk of different VTE manifestations (deep venous thrombosis, pulmonary embolism, pregnancy-related VTE, unusual thrombosis, and superficial thrombophlebitis), thrombophilia, and lower age of first VTE event. Moreover, VTE density in pedigrees was significantly associated in the offspring with obesity, diabetes, gout, varicose veins, and arterial embolism and thrombosis (excluding brain and heart). No significant associations were observed for retinal vein occlusion, hypercholesterolemia, hypertension, coronary heart disease, myocardial infarction, ischemic stroke, atrial fibrillation, heart failure, primary pulmonary hypertension, cerebral hemorrhage, aortic aneurysm, peripheral artery disease, and overall mortality. CONCLUSION Offspring of pedigrees with a high density of VTE are disadvantaged regarding VTE manifestations and certain cardiometabolic disorders.
Collapse
Affiliation(s)
- Bengt Zöller
- Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden.
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden
| | - Henrik Ohlsson
- Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden
| |
Collapse
|
34
|
Sacchetti S, Puricelli C, Mennuni M, Zanotti V, Giacomini L, Giordano M, Dianzani U, Patti G, Rolla R. Research into New Molecular Mechanisms in Thrombotic Diseases Paves the Way for Innovative Therapeutic Approaches. Int J Mol Sci 2024; 25:2523. [PMID: 38473772 DOI: 10.3390/ijms25052523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/12/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Thrombosis is a multifaceted process involving various molecular components, including the coagulation cascade, platelet activation, platelet-endothelial interaction, anticoagulant signaling pathways, inflammatory mediators, genetic factors and the involvement of various cells such as endothelial cells, platelets and leukocytes. A comprehensive understanding of the molecular signaling pathways and cell interactions that play a role in thrombosis is essential for the development of precise therapeutic strategies for the treatment and prevention of thrombotic diseases. Ongoing research in this field is constantly uncovering new molecular players and pathways that offer opportunities for more precise interventions in the clinical setting. These molecular insights into thrombosis form the basis for the development of targeted therapeutic approaches for the treatment and prevention of thrombotic disease. The aim of this review is to provide an overview of the pathogenesis of thrombosis and to explore new therapeutic options.
Collapse
Affiliation(s)
- Sara Sacchetti
- Clinical Chemistry Laboratory, "Maggiore della Carità" University Hospital, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
| | - Chiara Puricelli
- Clinical Chemistry Laboratory, "Maggiore della Carità" University Hospital, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
| | - Marco Mennuni
- Division of Cardiology, "Maggiore della Carità" University Hospital, Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy
| | - Valentina Zanotti
- Clinical Chemistry Laboratory, "Maggiore della Carità" University Hospital, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
| | - Luca Giacomini
- Clinical Chemistry Laboratory, "Maggiore della Carità" University Hospital, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
| | - Mara Giordano
- Clinical Chemistry Laboratory, "Maggiore della Carità" University Hospital, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
| | - Umberto Dianzani
- Clinical Chemistry Laboratory, "Maggiore della Carità" University Hospital, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
| | - Giuseppe Patti
- Division of Cardiology, "Maggiore della Carità" University Hospital, Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy
| | - Roberta Rolla
- Clinical Chemistry Laboratory, "Maggiore della Carità" University Hospital, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
| |
Collapse
|
35
|
Han J, Li‐Gao R, de Mutsert R, Rosendaal FR, van Hylckama Vlieg A. Association between venous thromboembolism-associated genetic variants, coagulation factor levels, and thrombin generation potential. EJHAEM 2024; 5:47-54. [PMID: 38406509 PMCID: PMC10887265 DOI: 10.1002/jha2.863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/27/2024]
Abstract
Recently three large meta-analyses of genome-wide association studies for venous thromboembolism (VTE) identified over 130 genetic variants. However, mechanisms by which newly identified and therefore underexplored VTE-associated genetic variants influence VTE remain unclear. To elucidate the mechanism, we investigated the association between 61 newly identified VTE-associated genetic variants and the levels of coagulation factor (F) VIII, FIX, FXI, and fibrinogen as well as thrombin generation parameters (lag time, peak, endogenous thrombin potential, time-to-peak, and velocity), which are well-known biological traits associated with VTE. This study was conducted on 5341 participants of the Netherlands Epidemiology of Obesity study. The associations between VTE-associated genetic variants and coagulation factor levels and thrombin generation parameters were examined using linear regression analyses, adjusted for age, sex, body mass index, oral contraceptive use, hormone replacement therapy, and menopausal status. Of 61 genetic variants, 33 were associated with one or more of the coagulation factor levels and thrombin generation parameters. Following multiple testing corrections, five genetic variants remained significant, of which MAP1A rs55707100 exhibited the most robust association with thrombin generation parameters and FXI levels (β = -5.33%, 95% confidence interval: -8.44, -2.22). Our findings shed light on the underlying mechanisms by which these genetic variants influence the risk of VTE.
Collapse
Affiliation(s)
- Jihee Han
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Ruifang Li‐Gao
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Renée de Mutsert
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Frits R. Rosendaal
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenthe Netherlands
| | | |
Collapse
|
36
|
Løchen Arnesen CA, Evensen LH, Hveem K, Gabrielsen ME, Hansen JB, Brækkan SK. Proportion of venous thromboembolism attributed to recognized prothrombotic genotypes in men and women. Res Pract Thromb Haemost 2024; 8:102343. [PMID: 38476459 PMCID: PMC10926210 DOI: 10.1016/j.rpth.2024.102343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 03/14/2024] Open
Abstract
Background Data on the proportion of venous thromboembolism (VTE) risk attributed to prothrombotic genotypes in men and women are limited. Objectives We aimed to estimate the population attributable fraction (PAF) of VTE for recognized, common prothrombotic genotypes in men and women using a population-based case cohort. Methods Cases with incident VTE (n = 1493) and a randomly sampled subcohort (n = 13,069) were derived from the Tromsø study (1994-2012) and the Trøndelag Health Study (1995-2008) cohorts. DNA samples were genotyped for 17 single-nucleotide polymorphisms (SNPs) previously associated with VTE. PAFs with 95% bias-corrected CIs (based on 10,000 bootstrap samples) were estimated for SNPs significantly associated with VTE, and a 6-SNP cumulative model was constructed for both sexes. Results In women, the individual PAFs for SNPs included in the cumulative model were 16.9% for ABO (rs8176719), 17.6% for F11 (rs2036914), 15.1% for F11 (rs2289252), 8.7% for FVL (rs6025), 6.0% for FGG (rs2066865), and 0.2% for F2 (rs1799963). The cumulative PAF for this 6-SNP model was 37.8%. In men, the individual PAFs for SNPs included in the cumulative model were 21.3% for ABO, 12.2% for F11 (rs2036914), 10.4% for F11 (rs2289252), 7.5% for FVL, 7.8% for FGG, and 1.1% for F2. This resulted in a cumulative PAF in men of 51.9%. Conclusion Our findings in a Norwegian population suggest that 52% and 38% of the VTEs can be attributed to known prothrombotic genotypes in men and women, respectively.
Collapse
Affiliation(s)
- Carl Arne Løchen Arnesen
- Thrombosis Research Group, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
- Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Line H Evensen
- Thrombosis Research Group, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Kristian Hveem
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Center, Department of Public Health and Nursing, Norwegian University of Science and Technology, Levanger, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Maiken E Gabrielsen
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - John-Bjarne Hansen
- Thrombosis Research Group, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
- Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Sigrid K Brækkan
- Thrombosis Research Group, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
- Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
37
|
Drăgan A, Drăgan AŞ. Novel Insights in Venous Thromboembolism Risk Assessment Methods in Ambulatory Cancer Patients: From the Guidelines to Clinical Practice. Cancers (Basel) 2024; 16:458. [PMID: 38275899 PMCID: PMC10813930 DOI: 10.3390/cancers16020458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/07/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Many cancer patients will experience venous thromboembolism (VTE) at some stage, with the highest rate in the initial period following diagnosis. Novel cancer therapies may further enhance the risk. VTE in a cancer setting is associated with poor prognostic, a decreased quality of life, and high healthcare costs. If thromboprophylaxis in hospitalized cancer patients and perioperative settings is widely accepted in clinical practice and supported by the guidelines, it is not the same situation in ambulatory cancer patient settings. The guidelines do not recommend primary thromboprophylaxis, except in high-risk cases. However, nowadays, risk stratification is still challenging, although many tools have been developed. The Khrorana score remains the most used method, but it has many limits. This narrative review aims to present the current relevant knowledge of VTE risk assessment in ambulatory cancer patients, starting from the guideline recommendations and continuing with the specific risk assessment methods and machine learning models approaches. Biomarkers, genetic, and clinical features were tested alone or in groups. Old and new models used in VTE risk assessment are exposed, underlining their clinical utility. Imaging and biomolecular approaches to VTE screening of outpatients with cancer are also presented, which could help clinical decisions.
Collapse
Affiliation(s)
- Anca Drăgan
- Department of Cardiovascular Anaesthesiology and Intensive Care, Emergency Institute for Cardiovascular Diseases “Prof. Dr. C C Iliescu”, 258 Fundeni Road, 022328 Bucharest, Romania
| | - Adrian Ştefan Drăgan
- Faculty of General Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania;
| |
Collapse
|
38
|
Zhu QM, Hsu YHH, Lassen FH, MacDonald BT, Stead S, Malolepsza E, Kim A, Li T, Mizoguchi T, Schenone M, Guzman G, Tanenbaum B, Fornelos N, Carr SA, Gupta RM, Ellinor PT, Lage K. Protein interaction networks in the vasculature prioritize genes and pathways underlying coronary artery disease. Commun Biol 2024; 7:87. [PMID: 38216744 PMCID: PMC10786878 DOI: 10.1038/s42003-023-05705-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/13/2023] [Indexed: 01/14/2024] Open
Abstract
Population-based association studies have identified many genetic risk loci for coronary artery disease (CAD), but it is often unclear how genes within these loci are linked to CAD. Here, we perform interaction proteomics for 11 CAD-risk genes to map their protein-protein interactions (PPIs) in human vascular cells and elucidate their roles in CAD. The resulting PPI networks contain interactions that are outside of known biology in the vasculature and are enriched for genes involved in immunity-related and arterial-wall-specific mechanisms. Several PPI networks derived from smooth muscle cells are significantly enriched for genetic variants associated with CAD and related vascular phenotypes. Furthermore, the networks identify 61 genes that are found in genetic loci associated with risk of CAD, prioritizing them as the causal candidates within these loci. These findings indicate that the PPI networks we have generated are a rich resource for guiding future research into the molecular pathogenesis of CAD.
Collapse
Affiliation(s)
- Qiuyu Martin Zhu
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Yu-Han H Hsu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Frederik H Lassen
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Bryan T MacDonald
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Stephanie Stead
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Edyta Malolepsza
- Genomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - April Kim
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Taibo Li
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Taiji Mizoguchi
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Monica Schenone
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gaelen Guzman
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Benjamin Tanenbaum
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nadine Fornelos
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Steven A Carr
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rajat M Gupta
- Divisions of Cardiovascular Medicine and Genetics, Brigham and Women's Hospital, Boston, MA, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| | - Kasper Lage
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA.
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark.
| |
Collapse
|
39
|
Jee YH, Thibord F, Dominguez A, Sept C, Boulier K, Venkateswaran V, Ding Y, Cherlin T, Verma SS, Faro VL, Bartz TM, Boland A, Brody JA, Deleuze JF, Emmerich J, Germain M, Johnson AD, Kooperberg C, Morange PE, Pankratz N, Psaty BM, Reiner AP, Smadja DM, Sitlani CM, Suchon P, Tang W, Trégouët DA, Zöllner S, Pasaniuc B, Damrauer SM, Sanna S, Snieder H, Kabrhel C, Smith NL, Kraft P. Multi-ancestry polygenic risk scores for venous thromboembolism. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.09.24300914. [PMID: 38260294 PMCID: PMC10802635 DOI: 10.1101/2024.01.09.24300914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Venous thromboembolism (VTE) is a significant contributor to morbidity and mortality, with large disparities in incidence rates between Black and White Americans. Polygenic risk scores (PRSs) limited to variants discovered in genome-wide association studies in European-ancestry samples can identify European-ancestry individuals at high risk of VTE. However, there is limited evidence on whether high-dimensional PRS constructed using more sophisticated methods and more diverse training data can enhance the predictive ability and their utility across diverse populations. We developed PRSs for VTE using summary statistics from the International Network against Venous Thrombosis (INVENT) consortium GWAS meta-analyses of European- (71,771 cases and 1,059,740 controls) and African-ancestry samples (7,482 cases and 129,975 controls). We used LDpred2 and PRSCSx to construct ancestry-specific and multi-ancestry PRSs and evaluated their performance in an independent European- (6,261 cases and 88,238 controls) and African-ancestry sample (1,385 cases and 12,569 controls). Multi-ancestry PRSs with weights tuned in European- and African-ancestry samples, respectively, outperformed ancestry-specific PRSs in European- (PRSCSXEUR: AUC=0.61 (0.60, 0.61), PRSCSX_combinedEUR: AUC=0.61 (0.60, 0.62)) and African-ancestry test samples (PRSCSXAFR: AUC=0.58 (0.57, 0.6), PRSCSX_combined AFR: AUC=0.59 (0.57, 0.60)). The highest fifth percentile of the best-performing PRS was associated with 1.9-fold and 1.68-fold increased risk for VTE among European- and African-ancestry subjects, respectively, relative to those in the middle stratum. These findings suggest that the multi-ancestry PRS may be used to identify individuals at highest risk for VTE and provide guidance for the most effective treatment strategy across diverse populations.
Collapse
Affiliation(s)
- Yon Ho Jee
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, MA, USA
| | - Florian Thibord
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, MD, USA
- The Framingham Heart Study, 73 Mt. Wayte Ave, Suite #2, Framingham, MA, 01702 USA
| | - Alicia Dominguez
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Corriene Sept
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Kristin Boulier
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Vidhya Venkateswaran
- Department of Oral Biology, University of California Los Angeles School of Dentistry, Los Angeles, CA, USA
| | - Yi Ding
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Tess Cherlin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shefali Setia Verma
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Valeria Lo Faro
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Traci M. Bartz
- Cardiovascular Health Research Unit, Departments of Biostatistics and Medicine, University of Washington, 4333 Brooklyn Ave, Seattle, WA 98195
| | - Anne Boland
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, Evry, France
- Laboratory of Excellence in Medical Genomics, GENMED, Evry, France
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, 4333 Brooklyn Ave, Seattle, WA 98195
| | - Jean-Francois Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, Evry, France
- Laboratory of Excellence in Medical Genomics, GENMED, Evry, France
- Centre d’Etude du Polymorphisme Humain, Fondation Jean Dausset, Paris, France
| | - Joseph Emmerich
- Department of Vascular Medicine, Paris Saint-Joseph Hospital Group, University of Paris, Paris, France
- UMR1153, INSERM CRESS, Paris, France
| | - Marine Germain
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR 1219, Bordeaux, France
| | - Andrew D. Johnson
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, MD, USA
- The Framingham Heart Study, 73 Mt. Wayte Ave, Suite #2, Framingham, MA, 01702 USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinbson Cancer Center, Seattle WA 98109
| | - Pierre-Emmanuel Morange
- Aix-Marseille University, INSERM, INRAE, Centre de Recherche en CardioVasculaire et Nutrition, Laboratory of Haematology, CRB Assistance Publique – Hôpitaux de Marseille, HemoVasc, Marseille, France
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, 55455, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, 4333 Brooklyn Ave, Seattle, WA 98195
- Department of Epidemiology, University of Washington, 4333 Brooklyn Ave, Seattle, WA 98195
- Department of Health Systems and Population Health, University of Washington, 4333 Brooklyn Ave, Seattle, WA 98195
| | - Alexander P. Reiner
- Division of Public Health Sciences, Fred Hutchinbson Cancer Center, Seattle WA 98109
- Department of Epidemiology, University of Washington, 4333 Brooklyn Ave, Seattle, WA 98195
| | - David M. Smadja
- Innovative Therapies in Hemostasis, Université de Paris, INSERM, F-75006 Paris, France
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), F-75015 Paris, France
| | - Colleen M. Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, 4333 Brooklyn Ave, Seattle, WA 98195
| | - Pierre Suchon
- Aix-Marseille University, INSERM, INRAE, Centre de Recherche en CardioVasculaire et Nutrition, Laboratory of Haematology, CRB Assistance Publique – Hôpitaux de Marseille, HemoVasc, Marseille, France
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, 55454, USA
| | - David-Alexandre Trégouët
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR 1219, Bordeaux, France
| | - Sebastian Zöllner
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Bogdan Pasaniuc
- Department of Oral Biology, University of California Los Angeles School of Dentistry, Los Angeles, CA, USA
| | - Scott M. Damrauer
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Surgery, Department of Genetics, and Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA
- Department of Surgery, Corporal Michael Crescenz VA Medical Center, Philadelphia PA
| | - Serena Sanna
- University of Groningen, UMCG, Department of Genetics, Groningen, the Netherlands
- Institute for Genetics and Biomedical Research, National Research Council, Monserrato, Italy
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Christopher Kabrhel
- Center for Vascular Emergencies, Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicholas L. Smith
- Department of Health Systems and Population Health, University of Washington, 4333 Brooklyn Ave, Seattle, WA 98195
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle WA 98101, USA
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle WA 98108, USA
| | - Peter Kraft
- Transdivisional Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, MD, USA
| | | |
Collapse
|
40
|
Liu C, Hou J, Li W, Chen J, Li Y, Zhang J, Zhou W, Zhang W, Deng F, Wang Y, Chen L, Qin S, Meng X, Lu S. Construction and optimization of a polygenic risk model for venous thromboembolism in the Chinese population. J Vasc Surg Venous Lymphat Disord 2024; 12:101666. [PMID: 37619711 PMCID: PMC11523315 DOI: 10.1016/j.jvsv.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/06/2023] [Accepted: 08/12/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND Venous thromboembolism (VTE) has both environmental and genetic risk factors. It is regulated by polygenes and multisites. The polygenic risk score (PRS) has been widely used because any single genetic biomarker failed to accurately predict the genetic risk of VTE. However, no polygenic risk model has been proposed for VTE in the Chinese population. Thus, we aimed to construct a PRS model for the first episode of VTE in the Chinese population. METHODS First, single nucleotide polymorphisms (SNPs) associated with VTE in genome-wide association studies, meta-analyses, and candidate gene studies were screened as variables for the PRS. The logarithm of the odds ratio was used to weight the variables. Second, a training set with simulated data from 1000 cases of VTE and 1000 controls was created with different genotypes and frequencies. Finally, we calculated the area under the receiver operating characteristic curve (AUC) to evaluate the discriminatory ability of the PRS model. RESULTS We screened 53 SNPs potentially associated with the first episode of VTE in the Chinese population. The AUC of the PRS-53 model (containing 53 SNPs) was 0.748 (95% confidence interval, 0.727-0.770) in the training set. From the largest weight to the smallest weight, SNPs were incrementally added to the model to calculate the AUC for model optimization. The AUC of the PRS-10 model (containing 10 SNPs) was 0.718 (95% confidence interval, 0.696-0.740), with no statistically significant difference from the AUC for the PRS-53 model. CONCLUSIONS The PRS-10 and PRS-53 models showed similar predictive abilities and satisfactory discriminatory power and can be used to predict the genetic risk of the first episode of VTE in the Chinese population. The simplified PRS-10 model is more efficient in clinical practice.
Collapse
Affiliation(s)
- Chao Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, People's Republic of China
| | - Jiaxuan Hou
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, People's Republic of China
| | - Weiming Li
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, People's Republic of China
| | - Jinxing Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, People's Republic of China
| | - Yane Li
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, People's Republic of China
| | - Jiawei Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, People's Republic of China
| | - Wei Zhou
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Wei Zhang
- Xi'an Agen Medicine Technology Co, Ltd, Xi'an, People's Republic of China
| | - Fenni Deng
- Xi'an Agen Medicine Technology Co, Ltd, Xi'an, People's Republic of China
| | - Yu Wang
- Xi'an Agen Medicine Technology Co, Ltd, Xi'an, People's Republic of China
| | - Luan Chen
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Shengying Qin
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Xiaohong Meng
- Xi'an Agen Medicine Technology Co, Ltd, Xi'an, People's Republic of China
| | - Shaoying Lu
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, People's Republic of China.
| |
Collapse
|
41
|
Jan K, Pavel J, Renata C, Daniela D, Petra B, Martin Š, Zuzana Z, Tomáš K. The incidence of the thrombophilic SNPs rs6025, rs1799963, rs2066865, rs2289252, and rs8176719 in chronic thromboembolic pulmonary hypertension. Clin Appl Thromb Hemost 2024; 30:10760296241271369. [PMID: 39150410 PMCID: PMC11329958 DOI: 10.1177/10760296241271369] [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: 04/15/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 08/17/2024] Open
Abstract
INTRODUCTION Chronic thromboembolic pulmonary hypertension (CTEPH) and venous thromboembolism (VTE) are thought to share many common risk factors. Our study aimed to determine the frequencies of 5 thrombosis-related gene single nucleotide polymorphisms (SNPs) associated with VTE in patients with CTEPH (n 129) compared with a control group of healthy individuals without a history of VTE (n 2637). METHODS The SNPs of the following genes were investigated: F5 (F V Leiden, rs6025), F2 prothrombin (rs1799963), fibrinogen gamma (FGG, rs2066865), F11 (rs2289252) and ABO (non-O, rs8176719) in both groups. RESULTS The study found that the rs1799963 variant was more common in patients with chronic thromboembolic pulmonary hypertension (CTEPH) compared to the control group (p < .0001). The GA heterozygous variant showed a significant increase with an odds ratio (OR) of 4.480 (95% CI: 2.344-8.562) or a finding by maximum likelihood analysis (MLA) with p < .0001. Additionally, there was a notable increase in the rs8176719 variant with p < .0001 in CTEPH patients. Both the homozygous G/G variant and the heterozygous -/G variant also showed an increase, with OR of 4.2317 (95% CI: 2.45571-7.2919) and 2.4324 (95% CI: 1.46435-4.0403) respectively, or MLA (p < .0001 and p .0006). The study also revealed a higher prevalence of the heterozygous C/T variant of rs2289252 in CTEPH patients, with an OR of 1.5543 (95% CI: 1.02503-2.3568) or MLA (p .0379). CONCLUSION The study suggests that the observed gene polymorphisms F2 (rs1799963), ABO (rs8176719), and F11 (rs2289252) may play a role as independent heritable risk factors in the development of CTEPH.
Collapse
Affiliation(s)
- Kvasnička Jan
- Thrombotic Centre, Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
- 1st Department of Medicine, Division of Haematology, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Jansa Pavel
- 2nd Department of Medicine, Division of Cardiovascular Medicine, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Cífková Renata
- Centre for Cardiovascular Prevention, 1st Faculty of Medicine, Charles University and Thomayer University Hospital, Prague, Czech Republic
| | - Dušková Daniela
- 1st Department of Medicine, Division of Haematology, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
- Department of Blood Transfusion, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Bobčíková Petra
- Thrombotic Centre, Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Ševčík Martin
- Thrombotic Centre, Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Zenáhlíková Zuzana
- Thrombotic Centre, Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Kvasnička Tomáš
- Thrombotic Centre, Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| |
Collapse
|
42
|
LOWENSTEIN CHARLESJ. GENETIC DETERMINANTS OF THROMBOSIS. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2024; 134:230-238. [PMID: 39135563 PMCID: PMC11316864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Venous thromboembolism (VTE) is a major cause of morbidity and mortality in the United States. VTE is caused by genetic and acquired conditions, but the genetic variants that increase the risk of VTE are not fully characterized. Recent genome-wide association studies (GWAS) have discovered novel genetic loci linked to VTE. Some of these loci have been characterized, uncovering new pathways that regulate VTE. Functional characterization of candidate genes discovered by GWAS may reveal new therapeutic targets to treat and prevent abnormal thrombosis or bleeding.
Collapse
|
43
|
He C, Chu J, Li Y, Dang Y, Xue K, Cai C. Reassessing the Risk of Venous Thromboembolism (VTE) Events in Women. Clin Appl Thromb Hemost 2024; 30:10760296241305108. [PMID: 39648738 PMCID: PMC11626654 DOI: 10.1177/10760296241305108] [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: 10/01/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 12/10/2024] Open
Abstract
This research aims to reassess women's risk of venous thromboembolism (VTE) events. We conducted an in-depth analysis of the environmental risk factors associated with VTE and their interactions with gender while also exploring the genetic underpinnings of the disease. VTE is identified as a multifactorial condition influenced by a combination of genetic, non-predisposing, and predisposing environmental factors. We further investigated the genetic basis of VTE, focusing on the identification and analysis of risk loci, as well as gene interaction networks and genetic analyses, which offer significant insights into the pathogenesis of VTE. Recognizing the critical role of gender in assessing VTE risk and developing prevention strategies, this research underscores the necessity of adopting an integrated perspective that accounts for individual vulnerabilities at both genetic and environmental levels to formulate effective preventive measures.
Collapse
Affiliation(s)
- Changhuai He
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Jie Chu
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiping Dang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kaming Xue
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuanqi Cai
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
44
|
Hasbani NR, Westerman KE, Kwak SH, Chen H, Li X, Di Corpo D, Wessel J, Bis JC, Sarnowski C, Wu P, Bielak LF, Guo X, Heard-Costa N, Kinney GL, Mahaney MC, Montasser ME, Palmer ND, Raffield LM, Terry JG, Yanek LR, Bon J, Bowden DW, Brody JA, Duggirala R, Jacobs DR, Kalyani RR, Lange LA, Mitchell BD, Smith JA, Taylor KD, Carson AP, Curran JE, Fornage M, Freedman BI, Gabriel S, Gibbs RA, Gupta N, Kardia SLR, Kral BG, Momin Z, Newman AB, Post WS, Viaud-Martinez KA, Young KA, Becker LC, Bertoni AG, Blangero J, Carr JJ, Pratte K, Psaty BM, Rich SS, Wu JC, Malhotra R, Peyser PA, Morrison AC, Vasan RS, Lin X, Rotter JI, Meigs JB, Manning AK, de Vries PS. Type 2 Diabetes Modifies the Association of CAD Genomic Risk Variants With Subclinical Atherosclerosis. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:e004176. [PMID: 38014529 PMCID: PMC10843644 DOI: 10.1161/circgen.123.004176] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 09/29/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Individuals with type 2 diabetes (T2D) have an increased risk of coronary artery disease (CAD), but questions remain about the underlying pathology. Identifying which CAD loci are modified by T2D in the development of subclinical atherosclerosis (coronary artery calcification [CAC], carotid intima-media thickness, or carotid plaque) may improve our understanding of the mechanisms leading to the increased CAD in T2D. METHODS We compared the common and rare variant associations of known CAD loci from the literature on CAC, carotid intima-media thickness, and carotid plaque in up to 29 670 participants, including up to 24 157 normoglycemic controls and 5513 T2D cases leveraging whole-genome sequencing data from the Trans-Omics for Precision Medicine program. We included first-order T2D interaction terms in each model to determine whether CAD loci were modified by T2D. The genetic main and interaction effects were assessed using a joint test to determine whether a CAD variant, or gene-based rare variant set, was associated with the respective subclinical atherosclerosis measures and then further determined whether these loci had a significant interaction test. RESULTS Using a Bonferroni-corrected significance threshold of P<1.6×10-4, we identified 3 genes (ATP1B1, ARVCF, and LIPG) associated with CAC and 2 genes (ABCG8 and EIF2B2) associated with carotid intima-media thickness and carotid plaque, respectively, through gene-based rare variant set analysis. Both ATP1B1 and ARVCF also had significantly different associations for CAC in T2D cases versus controls. No significant interaction tests were identified through the candidate single-variant analysis. CONCLUSIONS These results highlight T2D as an important modifier of rare variant associations in CAD loci with CAC.
Collapse
Affiliation(s)
- Natalie R Hasbani
- Department of Epidemiology Human Genetics and Environmental Sciences, Human Genetics Center, The University of Texas Health Science Center at Houston School of Public Health (N.R.H., H.C., C.S., A.C.M., P.S.d.V.)
| | - Kenneth E Westerman
- Department of Medicine, Clinical and Translation Epidemiology Unit (K.E.W., A.K.M.), Massachusetts General Hospital, Boston
- Programs in Metabolism and Medical and Population Genetics (K.E.W., J.B.M., A.K.M.), Broad Institute, Cambridge
- Department of Medicine, Harvard Medical School, Boston, MA (K.E.W., J.B.M., A.K.M.)
| | - Soo Heon Kwak
- Department of Internal Medicine, Seoul National University Hospital, South Korea (S.H.K.)
| | - Han Chen
- Department of Epidemiology Human Genetics and Environmental Sciences, Human Genetics Center, The University of Texas Health Science Center at Houston School of Public Health (N.R.H., H.C., C.S., A.C.M., P.S.d.V.)
- School of Biomedical Informatics, Center for Precision Health (H.C.), The University of Texas Health Science Center at Houston
| | - Xihao Li
- Department of Biostatistics, Harvard T.H. Chan School of Public Health (X. Li, X. Lin), Boston University School of Public Health, MA
| | - Daniel Di Corpo
- Department of Biostatistics (D.D., P.W.), Boston University School of Public Health, MA
| | - Jennifer Wessel
- Department of Epidemiology, Fairbanks School of Public Health, Indianapolis, IN (J.W.)
| | - Joshua C Bis
- Department of Medicine, Cardiovascular Health Research Unit (J.C.B., J.A.B., B.M.P.), University of Washington, Seattle
| | - Chloè Sarnowski
- Department of Epidemiology Human Genetics and Environmental Sciences, Human Genetics Center, The University of Texas Health Science Center at Houston School of Public Health (N.R.H., H.C., C.S., A.C.M., P.S.d.V.)
| | - Peitao Wu
- Department of Biostatistics (D.D., P.W.), Boston University School of Public Health, MA
| | - Lawrence F Bielak
- Department of Medicine, Harvard Medical School, Boston, MA (K.E.W., J.B.M., A.K.M.)
| | - Xiuqing Guo
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, Torrance (X.G., K.D.T.)
| | | | - Gregory L Kinney
- Department of Epidemiology, University of Colorado School of Public Health, Aurora (G.L.K., K.A.Y.)
| | - Michael C Mahaney
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville (M.C.M., J.E.C., J. Blangero)
| | - May E Montasser
- Department of Medicine, Division of Endocrinology Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore (M.E.M., B.D.M.)
| | - Nicholette D Palmer
- Department of Biochemistry (N.D.P., D.W.B.), Wake Forest School of Medicine, Winston-Salem, NC
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill (L.M.R.)
| | - James G Terry
- Department of Radiology, Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, Nashville, TN (J.G.T., J.J.C.)
| | - Lisa R Yanek
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (L.R.Y., R.R.K., B.G.K., L.C.B.)
| | - Jessica Bon
- Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, PA (J. Bon)
| | - Donald W Bowden
- Department of Biochemistry (N.D.P., D.W.B.), Wake Forest School of Medicine, Winston-Salem, NC
| | - Jennifer A Brody
- Department of Medicine, Cardiovascular Health Research Unit (J.C.B., J.A.B., B.M.P.), University of Washington, Seattle
| | - Ravindranath Duggirala
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, McAllen (R.D.)
| | | | - Rita R Kalyani
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (L.R.Y., R.R.K., B.G.K., L.C.B.)
| | - Leslie A Lange
- Division of Biomedical Informatics and Personalized Medicine, School of Medicine University of Colorado, Aurora (L.A.L.)
| | - Braxton D Mitchell
- Department of Medicine, Division of Endocrinology Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore (M.E.M., B.D.M.)
- Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, MD (B.D.M.)
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor (L.F.B., J.A.S., S.L.R.K., P.A.P.)
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor (J.A.S.)
| | - Kent D Taylor
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, Torrance (X.G., K.D.T.)
| | - April P Carson
- Department of Medicine, University of Mississippi Medical Center, Jackson (A.P.C.)
| | - Joanne E Curran
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville (M.C.M., J.E.C., J. Blangero)
| | - Myriam Fornage
- Institute of Molecular Medicine (M.F.), The University of Texas Health Science Center at Houston
| | - Barry I Freedman
- Department of Internal Medicine, Section on Nephrology (B.I.F.), Wake Forest School of Medicine, Winston-Salem, NC
| | - Stacey Gabriel
- Genomics Platform (S.G., N.G.), Broad Institute, Cambridge
| | - Richard A Gibbs
- Baylor College of Medicine Human Genome Sequencing Center, Houston, TX (R.A.G., Z.M.)
| | - Namrata Gupta
- Genomics Platform (S.G., N.G.), Broad Institute, Cambridge
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor (L.F.B., J.A.S., S.L.R.K., P.A.P.)
| | - Brian G Kral
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (L.R.Y., R.R.K., B.G.K., L.C.B.)
| | - Zeineen Momin
- Baylor College of Medicine Human Genome Sequencing Center, Houston, TX (R.A.G., Z.M.)
| | - Anne B Newman
- Department of Epidemiology, University of Pittsburgh School of Public Health, PA (A.B.N.)
| | - Wendy S Post
- Division of Cardiology, Johns Hopkins Medicine, Baltimore, MD (W.S.P.)
| | | | - Kendra A Young
- Department of Epidemiology, University of Colorado School of Public Health, Aurora (G.L.K., K.A.Y.)
| | - Lewis C Becker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (L.R.Y., R.R.K., B.G.K., L.C.B.)
| | - Alain G Bertoni
- Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC (A.G.B.)
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville (M.C.M., J.E.C., J. Blangero)
| | - John J Carr
- Department of Radiology, Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, Nashville, TN (J.G.T., J.J.C.)
| | - Katherine Pratte
- Department of Biostatistics, National Jewish Health, Denver, CO (K.P.)
| | - Bruce M Psaty
- Department of Medicine, Cardiovascular Health Research Unit (J.C.B., J.A.B., B.M.P.), University of Washington, Seattle
- Department of Epidemiology (B.M.P.), University of Washington, Seattle
- Department of Health Systems and Population Health (B.M.P.), University of Washington, Seattle
| | | | - Joseph C Wu
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville (J.C.W.)
- Department of Medicine, Division of Cardiovascular Medicine, Stanford Cardiovascular Institute, Stanford University School of Medicine (J.C.W.), Stanford University, CA
| | - Rajeev Malhotra
- Division of Cardiology (R.M.), Massachusetts General Hospital, Boston
- Department of Radiology Molecular Imaging Program at Stanford (R.M.), Stanford University, CA
| | - Patricia A Peyser
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor (L.F.B., J.A.S., S.L.R.K., P.A.P.)
| | - Alanna C Morrison
- Department of Epidemiology Human Genetics and Environmental Sciences, Human Genetics Center, The University of Texas Health Science Center at Houston School of Public Health (N.R.H., H.C., C.S., A.C.M., P.S.d.V.)
| | - Ramachandran S Vasan
- Framingham Heart Study, MA (N.H.-C., R.S.V.)
- Department of Quantitative and Qualitative Health Sciences, University of Texas Health San Antonio School of Public Health (R.S.V.)
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health (X. Li, X. Lin), Boston University School of Public Health, MA
| | | | - James B Meigs
- Division of General Internal Medicine (J.B.M.), Massachusetts General Hospital, Boston
- Programs in Metabolism and Medical and Population Genetics (K.E.W., J.B.M., A.K.M.), Broad Institute, Cambridge
- Department of Medicine, Harvard Medical School, Boston, MA (K.E.W., J.B.M., A.K.M.)
| | - Alisa K Manning
- Department of Medicine, Clinical and Translation Epidemiology Unit (K.E.W., A.K.M.), Massachusetts General Hospital, Boston
- Programs in Metabolism and Medical and Population Genetics (K.E.W., J.B.M., A.K.M.), Broad Institute, Cambridge
- Department of Medicine, Harvard Medical School, Boston, MA (K.E.W., J.B.M., A.K.M.)
| | - Paul S de Vries
- Department of Epidemiology Human Genetics and Environmental Sciences, Human Genetics Center, The University of Texas Health Science Center at Houston School of Public Health (N.R.H., H.C., C.S., A.C.M., P.S.d.V.)
| |
Collapse
|
45
|
Han J, van Hylckama Vlieg A, Rosendaal FR. Genomic science of risk prediction for venous thromboembolic disease: convenient clarification or compounding complexity. J Thromb Haemost 2023; 21:3292-3303. [PMID: 37838557 DOI: 10.1016/j.jtha.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 09/07/2023] [Indexed: 10/16/2023]
Abstract
Venous thromboembolism (VTE) refers to abnormal blood clots in veins occurring in 1 to 2 per 1000 individuals every year. While anticoagulant treatment can prevent VTE, it increases the risk of bleeding. This emphasizes the importance of identifying individuals with a high risk of VTE and providing prophylactic interventions to these individuals to reduce both VTE and bleeding risks. Current risk assessment of VTE is based on the combination of mainly clinical risk factors. With the identification of an increasing number of genetic variants associated with the risk of VTE, the addition of genetic findings to clinical prediction models can improve risk prediction for VTE. Especially for individuals in high-risk situations, the added value of genetic findings to clinical prediction models may have benefits such as better prophylaxis of VTE and the reduced side effects of bleeding from unnecessary treatment. Nevertheless, the question of whether these models will eventually have clinical utility remains to be proven. Here, we review the current state of knowledge on genetic risk factors for VTE, explore genetic prediction models for VTE, and discuss their clinical implications and challenges.
Collapse
Affiliation(s)
- Jihee Han
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
46
|
Han X, Knauss EA, de la Fuente M, Li W, Conlon RA, LePage DF, Jiang W, Renna SA, McKenzie SE, Nieman MT. A Mouse Model of the Protease Activated Receptor 4 (PAR4) Pro310Leu Variant has Reduced Platelet Reactivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569075. [PMID: 38077081 PMCID: PMC10705540 DOI: 10.1101/2023.12.01.569075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Background Protease activated receptor 4 (PAR4) mediates thrombin signaling on platelets and other cells. Our recent structural studies demonstrated a single nucleotide polymorphism in extracellular loop 3 (ECL3), PAR4-P310L (rs2227376) leads to a hypo-reactive receptor. Objectives The goal of this study was to determine how the hypo-reactive PAR4 variant in ECL3 impacts platelet function in vivo using a novel knock-in mouse model (PAR4-322L). Methods A point mutation was introduced into the PAR4 gene, F2rl3, via CRISPR/Cas9 to create PAR4-P322L, the mouse homolog to human PAR4-P310L. Platelet response to PAR4 activation peptide (AYPGKF), thrombin, ADP, and convulxin was monitored by αIIbβ3 integrin activation and P-selectin translocation using flow cytometry or platelet aggregation. In vivo responses were determined by the tail bleeding assay and the ferric chloride-induced carotid artery injury model. Results PAR4-P/L and PAR4-L/L platelets had a reduced response to AYPGKF and thrombin measured by P-selectin translocation or αIIbβ3 activation. The response to ADP and convulxin was unchanged among genotypes. In addition, both PAR4-P/L and PAR4-L/L platelets showed a reduced response to thrombin in aggregation studies. There was an increase in the tail bleeding time for PAR4-L/L mice. The PAR4-P/L and PAR4-L/L mice both showed an extended time to arterial thrombosis. Conclusions PAR4-322L significantly reduced platelet responsiveness to AYPGKF and thrombin, which is in agreement with our previous structural and cell signaling studies. In addition, PAR4-322L had prolonged arterial thrombosis time. Our mouse model provides a foundation to further evaluate the role of PAR4 in other pathophysiological contexts.
Collapse
Affiliation(s)
- Xu Han
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH United States
| | - Elizabeth A. Knauss
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH United States
| | - Maria de la Fuente
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH United States
| | - Wei Li
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV United States
| | - Ronald A Conlon
- Case Transgenic and Targeting Facility, Case Western Reserve University, Cleveland, OH United States
| | - David F. LePage
- Case Transgenic and Targeting Facility, Case Western Reserve University, Cleveland, OH United States
| | - Weihong Jiang
- Case Transgenic and Targeting Facility, Case Western Reserve University, Cleveland, OH United States
| | - Stephanie A. Renna
- Department of Medicine, The Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA United States
| | - Steven E. McKenzie
- Department of Medicine, The Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA United States
| | - Marvin T. Nieman
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH United States
| |
Collapse
|
47
|
Li H, Zhang Z, Qiu Y, Weng H, Yuan S, Zhang Y, Zhang Y, Xi L, Xu F, Ji X, Hao R, Yang P, Chen G, Zuo X, Zhai Z, Wang C. Proteome-wide mendelian randomization identifies causal plasma proteins in venous thromboembolism development. J Hum Genet 2023; 68:805-812. [PMID: 37537391 PMCID: PMC10678328 DOI: 10.1038/s10038-023-01186-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/19/2023] [Accepted: 07/23/2023] [Indexed: 08/05/2023]
Abstract
Genome-wide association studies (GWAS) have identified numerous risk loci for venous thromboembolism (VTE), but it is challenging to decipher the underlying mechanisms. We employed an integrative analytical pipeline to transform genetic associations to identify novel plasma proteins for VTE. Proteome-wide association studies (PWAS) were determined by functional summary-based imputation leveraging data from a genome-wide association analysis (14,429 VTE patients, 267,037 controls), blood proteomes (1348 cases), followed by Mendelian randomization, Bayesian colocalization, protein-protein interaction, and pathway enrichment analysis. Twenty genetically regulated circulating protein abundances (F2, F11, ABO, PLCG2, LRP4, PLEK, KLKB1, PROC, KNG1, THBS2, SERPINA1, RARRES2, CEL, GP6, SERPINE2, SERPINA10, OBP2B, EFEMP1, F5, and MSR1) were associated with VTE. Of these 13 proteins demonstrated Mendelian randomized correlations. Six proteins (F2, F11, PLEK, SERPINA1, RARRES2, and SERPINE2) had strong support in colocalization analysis. Utilizing multidimensional data, this study suggests PLEK, SERPINA1, and SERPINE2 as compelling proteins that may provide key hints for future research and possible diagnostic and therapeutic targets for VTE.
Collapse
Affiliation(s)
- Haobo Li
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhu Zhang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.
| | - Yuting Qiu
- Capital Medical University, Beijing, China
| | - Haoyi Weng
- WeGene, Shenzhen, China; Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, China
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yunxia Zhang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Yu Zhang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Capital Medical University, Beijing, China
| | - Linfeng Xi
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Capital Medical University, Beijing, China
| | - Feiya Xu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Capital Medical University, Beijing, China
| | - Xiaofan Ji
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Risheng Hao
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Capital Medical University, Beijing, China
| | - Peiran Yang
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Gang Chen
- WeGene, Shenzhen, China; Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, China
| | - Xianbo Zuo
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
| | - Zhenguo Zhai
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.
| | - Chen Wang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
48
|
Verdier H, Thomas P, Batista J, Kempster C, McKinney H, Gleadall N, Danesh J, Mumford A, Heemskerk JWM, Ouwehand WH, Downes K, Astle WJ, Turro E. A signature of platelet reactivity in CBC scattergrams reveals genetic predictors of thrombotic disease risk. Blood 2023; 142:1895-1908. [PMID: 37647652 PMCID: PMC10733829 DOI: 10.1182/blood.2023021100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/27/2023] [Accepted: 08/18/2023] [Indexed: 09/01/2023] Open
Abstract
Genetic studies of platelet reactivity (PR) phenotypes may identify novel antiplatelet drug targets. However, such studies have been limited by small sample sizes (n < 5000) because of the complexity of measuring PR. We trained a model to predict PR from complete blood count (CBC) scattergrams. A genome-wide association study of this phenotype in 29 806 blood donors identified 21 distinct associations implicating 20 genes, of which 6 have been identified previously. The effect size estimates were significantly correlated with estimates from a study of flow cytometry-measured PR and a study of a phenotype of in vitro thrombus formation. A genetic score of PR built from the 21 variants was associated with the incidence rates of myocardial infarction and pulmonary embolism. Mendelian randomization analyses showed that PR was causally associated with the risks of coronary artery disease, stroke, and venous thromboembolism. Our approach provides a blueprint for using phenotype imputation to study the determinants of hard-to-measure but biologically important hematological traits.
Collapse
Affiliation(s)
- Hippolyte Verdier
- Institut Pasteur, CNRS UMR 3751, Decision and Bayesian Computation, Université Paris Cité, Paris, France
| | - Patrick Thomas
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Joana Batista
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Carly Kempster
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Harriet McKinney
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Nicholas Gleadall
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Andrew Mumford
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
- South West National Health Service Genomic Medicine Service Alliance, Bristol, United Kingdom
| | | | - Willem H. Ouwehand
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Kate Downes
- Cambridge Genomics Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - William J. Astle
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Medical Research Council Biostatistics Unit, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, United Kingdom
| | - Ernest Turro
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
49
|
Guman NAM, Mulder FI, Ferwerda B, Zwinderman AH, Kamphuisen PW, Büller HR, van Es N. Polygenic risk scores for prediction of cancer-associated venous thromboembolism in the UK Biobank cohort study. J Thromb Haemost 2023; 21:3175-3183. [PMID: 37481074 DOI: 10.1016/j.jtha.2023.07.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/06/2023] [Accepted: 07/09/2023] [Indexed: 07/24/2023]
Abstract
BACKGROUND Guidelines recommend thromboprophylaxis for patients with cancer at high risk of venous thromboembolism (VTE). Polygenic risk scores may improve VTE prediction but have not yet been evaluated in patients with cancer. OBJECTIVES We assessed the performance of the 5-, 37-, 297-, extended 297- (additionally including factor V Leiden and prothrombin G20210A), and 100-single-nucleotide polymorphism (SNP) scores in predicting cancer-associated VTE in the UK Biobank, a population-based, prospective cohort study. METHODS The primary outcome was VTE during 12 months after cancer diagnosis. Cancer and VTE diagnosis were based on ICD-10 codes. Discrimination was evaluated by c-indices and subdistribution hazard ratios in the upper vs 3 lower quartiles of the scores in a competing risk model. As a comparison, the c-index was calculated for the Khorana cancer type risk classification. RESULTS Of 36 150 patients with cancer (median age, 66 years; 48.7% females), 1018 (2.8%) developed VTE. C-indices at 12 months ranged from 0.56 (95% CI, 0.54-0.58) for the 5-SNP to 0.60 (95% CI, 0.58-0.62) for the extended 297-SNP scores. The subdistribution hazard ratios ranged from 1.36 (95% CI, 1.19-1.56) for the 5-SNP to 1.90 (95% CI, 1.68-2.16) for the extended 297-SNP scores and were consistent after adjusting for cancer type. For the Khorana cancer type classification, the c-index was 0.60 (95% CI, 0.58-0.61), which increased to 0.65 (95% CI, 0.63-0.67, +0.05; 95% CI, 0.04-0.07) when combined with the extended 297-SNP score. CONCLUSION These findings demonstrate that polygenic VTE risk scores can identify patients with cancer with a 1.9-fold higher VTE risk independent of cancer type. Combined clinical-genetic scores to improve cancer-associated VTE prediction should be evaluated further.
Collapse
Affiliation(s)
- Noori A M Guman
- Amsterdam UMC location University of Amsterdam, Vascular Medicine, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, The Netherlands; Department of Internal Medicine, Tergooi Medical Center, Hilversum, The Netherlands.
| | - Frits I Mulder
- Amsterdam UMC location University of Amsterdam, Vascular Medicine, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, The Netherlands; Department of Internal Medicine, Tergooi Medical Center, Hilversum, The Netherlands
| | - Bart Ferwerda
- Department of Clinical Epidemiology, Biostatistics, and Bioinformatics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Aeilko H Zwinderman
- Department of Clinical Epidemiology, Biostatistics, and Bioinformatics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Pieter W Kamphuisen
- Amsterdam UMC location University of Amsterdam, Vascular Medicine, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, The Netherlands; Department of Internal Medicine, Tergooi Medical Center, Hilversum, The Netherlands
| | - Harry R Büller
- Amsterdam UMC location University of Amsterdam, Vascular Medicine, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, The Netherlands
| | - Nick van Es
- Amsterdam UMC location University of Amsterdam, Vascular Medicine, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, The Netherlands
| |
Collapse
|
50
|
Alshabeeb MA, Alwadaani D, Al Qahtani FH, Abohelaika S, Alzahrani M, Al Zayed A, Al Saeed HH, Al Ajmi H, Alsomaie B, Rashid M, Daly AK. Impact of Genetic Variations on Thromboembolic Risk in Saudis with Sickle Cell Disease. Genes (Basel) 2023; 14:1919. [PMID: 37895268 PMCID: PMC10606407 DOI: 10.3390/genes14101919] [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: 07/16/2023] [Revised: 10/04/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Sickle cell disease (SCD) is a Mendelian disease characterized by multigenic phenotypes. Previous reports indicated a higher rate of thromboembolic events (TEEs) in SCD patients. A number of candidate polymorphisms in certain genes (e.g., FVL, PRT, and MTHFR) were previously reported as risk factors for TEEs in different clinical conditions. This study aimed to genotype these genes and other loci predicted to underlie TEEs in SCD patients. METHODOLOGY A multi-center genome-wide association study (GWAS) involving Saudi SCD adult patients with a history of TEEs (n = 65) and control patients without TEE history (n = 285) was performed. Genotyping used the 10× Affymetrix Axiom array, which includes 683,030 markers. Fisher's exact test was used to generate p-values of TEE associations with each single-nucleotide polymorphism (SNP). The haplotype analysis software tool version 1.05, designed by the University of Göttingen, Germany, was used to identify the common inherited haplotypes. RESULTS No association was identified between the targeted single-nucleotide polymorphism rs1801133 in MTHFR and TEEs in SCD (p = 0.79). The allele frequency of rs6025 in FVL and rs1799963 in PRT in our cohort was extremely low (<0.01); thus, both variants were excluded from the analysis as no meaningful comparison was possible. In contrast, the GWAS analysis showed novel genome-wide associations (p < 5 × 10-8) with seven signals; five of them were located on Chr 11 (rs35390334, rs331532, rs317777, rs147062602, and rs372091), one SNP on Chr 20 (rs139341092), and another on Chr 9 (rs76076035). The other 34 SNPs located on known genes were also detected at a signal threshold of p < 5 × 10-6. Seven of the identified variants are located in olfactory receptor family 51 genes (OR51B5, OR51V1, OR51A1P, and OR51E2), and five variants were related to family 52 genes (OR52A5, OR52K1, OR52K2, and OR52T1P). The previously reported association between rs5006884-A in OR51B5 and fetal hemoglobin (HbF) levels was confirmed in our study, which showed significantly lower levels of HbF (p = 0.002) and less allele frequency (p = 0.003) in the TEE cases than in the controls. The assessment of the haplotype inheritance pattern involved the top ten significant markers with no LD (rs353988334, rs317777, rs14788626882, rs49188823, rs139349992, rs76076035, rs73395847, rs1368823, rs8888834548, and rs1455957). A haplotype analysis revealed significant associations between two haplotypes (a risk, TT-AA-del-AA-ins-CT-TT-CC-CC-AA, and a reverse protective, CC-GG-ins-GG-del-TT-CC-TT-GG-GG) and TEEs in SCD (p = 0.024, OR = 6.16, CI = 1.34-28.24, and p = 0.019, OR = 0.33, CI = 0.13-0.85, respectively). CONCLUSIONS Seven markers showed novel genome-wide associations; two of them were exonic variants (rs317777 in OLFM5P and rs147062602 in OR51B5), and less significant associations (p < 5 × 10-6) were identified for 34 other variants in known genes with TEEs in SCD. Moreover, two 10-SNP common haplotypes were determined with contradictory effects. Further replication of these findings is needed.
Collapse
Affiliation(s)
- Mohammad A. Alshabeeb
- King Abdullah International Medical Research Center (KAIMRC), Riyadh 11426, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia (M.A.)
| | - Deemah Alwadaani
- King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia (M.A.)
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), Riyadh 11481, Saudi Arabia
| | - Farjah H. Al Qahtani
- Hematology/Oncology Center, King Saud University Medical City (KSUMC), Riyadh 11411, Saudi Arabia;
| | - Salah Abohelaika
- Research Department, Qatif Central Hospital (QCH), Qatif 32654, Saudi Arabia;
- Pharmacy Department, Qatif Central Hospital (QCH), Qatif 32654, Saudi Arabia
| | - Mohsen Alzahrani
- King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia (M.A.)
- King Fahad Hospital, Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
| | - Abdullah Al Zayed
- Hematology Department, Qatif Central Hospital (QCH), Qatif 32654, Saudi Arabia; (A.A.Z.); (H.H.A.S.)
| | - Hussain H. Al Saeed
- Hematology Department, Qatif Central Hospital (QCH), Qatif 32654, Saudi Arabia; (A.A.Z.); (H.H.A.S.)
| | - Hala Al Ajmi
- King Abdullah International Medical Research Center (KAIMRC), Riyadh 11426, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia (M.A.)
| | - Barrak Alsomaie
- King Abdullah International Medical Research Center (KAIMRC), Riyadh 11426, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia (M.A.)
| | - Mamoon Rashid
- King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia (M.A.)
- Department of AI and Bioinformatics, King Abdullah International Medical Research Center (KAIMRC), Riyadh 11481, Saudi Arabia
| | - Ann K. Daly
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK
| |
Collapse
|