1
|
Ganjibakhsh M, Tkachenko Y, Knutsen RH, Kozel BA. Toward a rational therapeutic for elastin related disease: Key considerations for elastin based regenerative medicine strategies. Matrix Biol 2025; 138:8-21. [PMID: 40158781 DOI: 10.1016/j.matbio.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/07/2025] [Accepted: 03/23/2025] [Indexed: 04/02/2025]
Abstract
Elastin is a connective tissue protein, produced from the ELN gene, that provides elasticity and recoil to tissues that stretch, such as the large arteries of the body, lung parenchyma, skin, ligaments and elastic cartilages. It is produced as a soluble monomer, tropoelastin, that when cross-linked in the extracellular space generates a polymer that is extraordinarily stable, with a predicted half-life of >70 years. Although data suggest ongoing elastin transcription, it is rare to see new elastin deposited outside of its tight developmental window. Consequently, elastin-related disease comes about primarily in one of three scenarios: (1) inadequate elastin deposition, (2) production of poor-quality elastic fibers, or (3) increased destruction of previously deposited elastin. By understanding the pathways controlling elastin production and maintenance, we can design new therapeutics to thwart those abnormal processes. In this review, we will summarize the diseases arising from genetic and environmental alteration of elastin (Williams syndrome, supravalvar aortic stenosis, autosomal dominant cutis laxa, and ELN-related vascular and connective tissue dysfunction) and then describe the mechanisms controlling elastin production and maintenance that might be manipulated to generate novel therapeutics aimed at these conditions. We will end by summarizing existing therapeutic strategies targeting these disease mechanisms before outlining future approaches that may better solve the challenges associated with elastin based regenerative medicine.
Collapse
Affiliation(s)
- Meysam Ganjibakhsh
- Institute of Genomic Medicine, Abigail Wexler Research Institute, Nationwide Children's Hospital, OH 43205, USA
| | - Yanina Tkachenko
- Institute of Genomic Medicine, Abigail Wexler Research Institute, Nationwide Children's Hospital, OH 43205, USA
| | - Russell H Knutsen
- Institute of Genomic Medicine, Abigail Wexler Research Institute, Nationwide Children's Hospital, OH 43205, USA
| | - Beth A Kozel
- Institute of Genomic Medicine, Abigail Wexler Research Institute, Nationwide Children's Hospital, OH 43205, USA; Department of Pediatrics, The Ohio State University, OH 43210, USA.
| |
Collapse
|
2
|
Berger T, Dumfarth J, Kreibich M, Minatoya K, Ziganshin BA, Czerny M. Thoracic aortic aneurysm. Nat Rev Dis Primers 2025; 11:34. [PMID: 40341396 DOI: 10.1038/s41572-025-00617-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/08/2025] [Indexed: 05/10/2025]
Abstract
Aortic medicine has undergone remarkable progress in recent decades with regard to our understanding and treatment of aortic disease. In the past decade, the scientific community has called for the aorta to be viewed as an independent organ, advocating for a holistic approach to understanding thoracic aortic disease, integrating its embryological development, wall composition, pathophysiological mechanisms, surveillance and treatment. Thoracic aortic aneurysm (TAA) is a potentially fatal disease characterized by abnormal dilation of the thoracic aorta, whereby the structural integrity of the vessel wall is compromised. Although epidemiological studies of TAA are confounded by its asymptomatic nature and diagnostic challenges, available evidence suggests that TAA prevalence and treatment outcomes vary according to race, sex and socioeconomic factors. Pathophysiological mechanisms involve interactions between vascular smooth muscle cells and the extracellular matrix, influenced by genetic predisposition and embryological factors as well as arterial hypertension. Diagnosis relies on advanced imaging techniques, with CT angiography considered to be the gold standard diagnostic tool and with genetic screening recommended for heritable conditions. Preventive measures focus on managing cardiovascular risk factors, whereas treatment includes medical management, as well as endovascular and open surgical repair. TAA has a major effect on quality of life, particularly in younger, female and genetically predisposed patients, necessitating further research and tailored interventions.
Collapse
Affiliation(s)
- Tim Berger
- Department of Cardiovascular Surgery, University Heart Center Freiburg - Bad Krozingen, University Medical Center Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Dumfarth
- University Clinic for Cardiac Surgery, Medical University Innsbruck, Innsbruck, Austria
| | - Maximilian Kreibich
- Department of Cardiovascular Surgery, University Heart Center Freiburg - Bad Krozingen, University Medical Center Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Bulat A Ziganshin
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT, USA
| | - Martin Czerny
- Department of Cardiovascular Surgery, University Heart Center Freiburg - Bad Krozingen, University Medical Center Freiburg, Freiburg, Germany.
- University Clinic for Cardiac Surgery, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
3
|
Teixido-Tura G, Dux-Santoy L, Badia C, Limeres J, Guala A, Evangelista Masip A, Ferreira-González I, Rodríguez-Palomares J. Present and future of aortic risk assessment in patients with heritable thoracic aortic diseases. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2025; 78:358-367. [PMID: 39536939 DOI: 10.1016/j.rec.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Heritable thoracic aortic diseases (HTAD) are a group of diverse genetic conditions characterized by an increased risk of aortic complications. The standard surveillance of these patients involves monitoring aortic diameters until a defined threshold is reached, at which point preventive aortic surgery is recommended. However, assessing aortic risk in these patients is far more complex and, in many aspects, remains incompletely understood. Several factors contribute to this complexity, including the diversity and low prevalence of the conditions within HTAD and the limited understanding of the factors influencing the progression of aortic dilation and the advent of acute aortic events. This article reviews current knowledge on clinical, genetic, and imaging factors related to aortic risk in HTAD and explores their potential future roles in improving risk assessment. By advancing our understanding of these factors, we aim to enhance the precision of risk stratification and develop more effective, personalized management strategies for HTAD patients, with the final goal of improving clinical outcomes and quality of life in individuals affected by these genetic disorders.
Collapse
Affiliation(s)
- Gisela Teixido-Tura
- Departamento de Cardiología, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Vall d'Hebron Institut de Recerca, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain.
| | | | - Clara Badia
- Departamento de Cardiología, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Javier Limeres
- Departamento de Cardiología, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Vall d'Hebron Institut de Recerca, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Andrea Guala
- Vall d'Hebron Institut de Recerca, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | | | - Ignacio Ferreira-González
- Departamento de Cardiología, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Vall d'Hebron Institut de Recerca, Barcelona, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain; Departamento de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - José Rodríguez-Palomares
- Departamento de Cardiología, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Vall d'Hebron Institut de Recerca, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; Departamento de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| |
Collapse
|
4
|
Yang W, Liu C, Li Z, Cui M. Exploring new drug treatment targets for immune related bone diseases using a multi omics joint analysis strategy. Sci Rep 2025; 15:10618. [PMID: 40148470 PMCID: PMC11950375 DOI: 10.1038/s41598-025-94053-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
In the field of treatment and prevention of immune-related bone diseases, significant challenges persist, necessitating the urgent exploration of new and effective treatment methods. However, most existing Mendelian randomization (MR) studies are confined to a single analytical approach, which limits the comprehensive understanding of the pathogenesis and potential therapeutic targets of these diseases. In light of this, we propose the hypothesis that genetic variations in specific plasma proteins have a causal relationship with immune-related bone diseases through the MR mechanism, and that key therapeutic targets can be accurately identified using an integrated multi-omic analysis approach. This study comprehensively applied a variety of analytical methods. Firstly, the protein quantitative trait locus (pQTLs) data from two large plasma protein databases and the Genome-Wide Association Study (GWAS) data of nine immune-related bone diseases were used for Mendelian randomization (MR) analysis. At the same time, we employed the Summary-based Mendelian Randomization (SMR) method, combined with the Bayesian colocalization analysis method of coding genes, as well as the Linkage Disequilibrium Score Regression (LDSC) analysis method based on genetic correlation analysis, as methods to verify the genetic association between genes and complex diseases, thus comprehensively obtaining positive results. In addition, a Phenome-wide Association Study (PheWAS) was conducted on significantly positive genes, and their expression patterns in different tissues were also explored. Subsequently, we integrated Protein-Protein Interaction (PPI) network analysis, Gene Ontology (GO) analysis. Finally, based on the above analytical methods, drug prediction and molecular docking studies were carried out with the aim of accurately identifying key therapeutic targets. Through a comprehensive analysis using four methods, namely the Mendelian randomization (MR) analysis study, Summary-based Mendelian Randomization (SMR) analysis study, Bayesian colocalization analysis study, and Linkage Disequilibrium Score Regression (LDSC) analysis study. We found that through MR, SMR, and combined with Bayesian colocalization analysis, an association was found between rheumatoid arthritis (RA) and HDGF. Using the combination of MR and Bayesian colocalization analysis, as well as LDSC analysis, it was concluded that RA was related to CCL19 and TNFRSF14. Based on the methods of MR and Bayesian colocalization, an association was found between GPT and Crohn's disease-related arthritis, and associations were found between BTN1A1, EVI5, OGA, TNFRSF14 and multiple sclerosis (MS), and associations were found between ICAM5, CCDC50, IL17RD, UBLCP1 and psoriatic arthritis (PsA). Specifically, in the MR analysis of RA, HDGF (P_ivw = 0.0338, OR = 1.0373, 95%CI = 1.0028-1.0730), CCL19 (P_ivw = 0.0004, OR = 0.3885, 95%CI = 0.2299-0.6566), TNFRSF14 (P_ivw = 0.0007, OR = 0.6947, 95%CI = 0.5634-0.8566); in the MR analysis of MS, BTN1A1 (P_ivw = 0.0000, OR = 0.6101, 95%CI = 0.4813-0.7733), EVI5 (P_ivw = 0.0000, OR = 0.3032, 95%CI = 0.1981-0.4642), OGA (P_ivw = 0.0005, OR = 0.4599, 95%CI = 0.2966-0.7131), TNFRSF14 (P_ivw = 0.0002, OR = 0.4026, 95%CI = 0.2505-0.6471); in the MR analysis of PsA, ICAM5 (P_ivw = 0.0281, OR = 1.1742, 95%CI = 1.0174-1.3552), CCDC50 (P_ivw = 0.0092, OR = 0.7359, 95%CI = 0.5843-0.9269), IL17RD (P_ivw = 0.0006, OR = 0.7887, 95%CI = 0.6886-0.9034), UBLCP1 (P_ivw = 0.0021, OR = 0.6901, 95%CI = 0.5448-0.8741); in the MR analysis of Crohn's disease-related arthritis, GPT (P_ivw = 0.0006, OR = 0.0057, 95%CI = 0.0003-0.1111). In the Bayesian colocalization analysis of RA, HDGF (H4 = 0.8426), CCL19 (H4 = 0.9762), TNFRSF14 (H4 = 0.8016); in the Bayesian colocalization analysis of MS, BTN1A1 (H4 = 0.7660), EVI5 (H4 = 0.9800), OGA (H4 = 0.8569), TNFRSF14 (H4 = 0.8904); in the Bayesian colocalization analysis of PsA, ICAM5 (H4 = 0.9476), CCDC50 (H4 = 0.9091), IL17RD (H4 = 0.9301), UBLCP1 (H4 = 0.8862); in the Bayesian colocalization analysis of Crohn's disease-related arthritis, GPT (H4 = 0.8126). In the SMR analysis of RA, HDGF (p_SMR = 0.0338, p_HEIDI = 0.0628). In the LDSC analysis of RA, CCL19 (P = 0.0000), TNFRSF14 (P = 0.0258). By comprehensively analyzing plasma proteomic and transcriptomic data, we successfully identified key therapeutic targets for various clinical subtypes of immune-associated bone diseases. Our findings indicate that the significant positive genes associated with RA include HDGF, CCL19, and TNFRSF14; the positive gene linked to Crohn-related arthropathy is GPT; for MS, the positive genes are BTN1A1, EVI5, OGA, and TNFRSF14; and for PsA, the positive genes are ICAM5, CCDC50, IL17RD, and UBLCP1. Through this comprehensive analytical approach, we have screened potential therapeutic targets for different clinical subtypes of immune-related bone diseases. This research not only enhances our understanding of the pathogenesis of these conditions but also provides a solid theoretical foundation for subsequent drug development and clinical treatment, with the potential to yield significant advancements in the management of patients with immune-related bone diseases.
Collapse
Affiliation(s)
- Wei Yang
- School of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Chenglin Liu
- School of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Zhenhua Li
- Affiliated Hospital of Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin, China.
| | - Miao Cui
- Capital Medical University, No.10, Xitoutiao, You'anmenwai, Beijing, 100069, Fengtai District, China.
| |
Collapse
|
5
|
Yang W, Liu C, Li Z, Cui M. Multi-omic biomarkers associated with multiple sclerosis: from Mendelian randomization to drug prediction. Sci Rep 2025; 15:9421. [PMID: 40108295 PMCID: PMC11923301 DOI: 10.1038/s41598-025-94303-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
Currently, the treatment and prevention of multiple sclerosis (MS) continue to encounter significant challenges. Mendelian randomization (MR) analysis has emerged as a crucial research method in the pursuit of new therapeutic strategies. Accordingly, we hypothesize that there exists a causal association between genetic variants of specific plasma proteins and MS through MR mechanisms, and that key therapeutic targets can be precisely identified by integrating multi-omics analytical approaches. In this study, we developed a comprehensive analytical framework aimed at identifying and validating potential therapeutic targets for MS. The framework commenced with a two-sample Mendelian randomization (MR) study utilizing two large plasma protein quantitative trait locus (pQTL) datasets. Building on this foundation, we performed Bayesian co-localization analysis of coding genes, followed by a full phenotype-wide association study (PheWAS) on the co-positive genes identified through both analytical methods. This approach allowed us to explore the functions of key genes and the mechanisms of co-morbidity associated with the disease. Subsequently, we integrated protein-protein interaction (PPI) network analysis, gene ontology (GO) analysis, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis to facilitate drug prediction and molecular docking studies. This study conducted a systematic analysis between two large plasma pQTLs datasets and MS. In the MR analysis, the MR analysis of Icelandic plasma pQTLs and MS identified 88 positive plasma proteins, while the MR analysis of the UK Biobank database pQTLs and MS identified 122 positive plasma proteins. By comparison, uroporphyrinogen III synthase (UROS) and glutathione S-transferase theta 2B (GSTT2B) were found to be the positive proteins shared by the two datasets. After false discovery rate (FDR) correction, signal transducer and activator of transcription 3 (STAT3) was a significantly positive protein in the analysis of Icelandic plasma pQTLs. In the analysis of the UK Biobank database pQTLs, advanced glycosylation end product-specific receptor (AGER), allograft inflammatory factor 1 (AIF1), butyrophilin subfamily 1 member A1 (BTN1A1), cluster of differentiation 58 (CD58), desmoglein 4 (DSG4), ecotropic viral integration site 5 (EVI5), tumor necrosis factor (TNF), and tumor necrosis factor receptor superfamily member 14 (TNFRSF14) were significantly positive proteins. After Bonferroni correction, AGER, CD58, EVI5, and TNF remained significantly positive proteins in the analysis of the UK Biobank database pQTLs. In the Bayesian colocalization analysis, EVI5 (PPH4 = 0.9800), O-GlcNAcase (OGA) (PPH4 = 0.8569), and TNFRSF14 (PPH4 = 0.8904) were the common positive genes in the two analysis methods. In conclusion, EVI5, OGA, and TNFRSF14 may be potential therapeutic targets for MS. Through the comprehensive application of MR analysis and Bayesian colocalization analysis, we have successfully identified that EVI5, OGA, and TNFRSF14 may be key therapeutic targets for MS. These findings may provide a scientific basis for the development of novel immunotherapies, combination treatment regimens, or targeted intervention strategies.
Collapse
Affiliation(s)
- Wei Yang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, China
| | - Chenglin Liu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, China
| | - Zhenhua Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, China.
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China.
| | - Miao Cui
- School of Traditional Chinese Medicine Fengtai District, Capital Medical University, No. 10, Xitoutiao Road, Beijing, 100069, China.
| |
Collapse
|
6
|
Bao M, Li H, Zhang Y, Li J. PM2.5 associates with blood pressure: a Mendelian randomization analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2024; 34:4190-4201. [PMID: 38647234 DOI: 10.1080/09603123.2024.2339536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 04/01/2024] [Indexed: 04/25/2024]
Abstract
The relationship between fine particulate matter (PM2.5) and blood pressure (BP) is a controversial issue. We conducted a two-sample Mendelian randomization (MR) analysis and identified 58 genome-wide significant single-nucleotide polymorphisms associated with PM2.5 as instrument variables. Inverse-variance weighted (IVW) was used as the primary analysis approach. MR-Egger, weighted median, simple model, and weighted model methods were selected for quality control. We found a significant negative causal association of higher genetically predicted PM2.5 levels with lower systolic BP (SBP), while no causal relationship was identified between PM2.5 and diastolic BP (DBP). For each 1 standard deviation increase in genetically predicted PM2.5 levels, the beta value (95% CI) of SBP was -0.14 (-0.25, -0.03) for IVW (p=0.02), and -0.13 (-0.22, -0.04) for weighted median (p=0.005). Increased PM2.5 concentrations can lead to decreased SBP levels. Our findings provided novel insights into the controversial topic on the causal relationship between PM2.5 and BP.
Collapse
Affiliation(s)
- Minghui Bao
- Department of Cardiology, Peking University First Hospital, Peking University, Beijing, China
| | - Haotong Li
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Zhang
- Department of Cardiology, Peking University First Hospital, Peking University, Beijing, China
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, Peking University, Beijing, China
| |
Collapse
|
7
|
Shine BK, Choi JE, Park YJ, Hong KW. The Genetic Variants Influencing Hypertension Prevalence Based on the Risk of Insulin Resistance as Assessed Using the Metabolic Score for Insulin Resistance (METS-IR). Int J Mol Sci 2024; 25:12690. [PMID: 39684400 DOI: 10.3390/ijms252312690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Insulin resistance is a major indicator of cardiovascular diseases, including hypertension. The Metabolic Score for Insulin Resistance (METS-IR) offers a simplified and cost-effective way to evaluate insulin resistance. This study aimed to identify genetic variants associated with the prevalence of hypertension stratified by METS-IR score levels. Data from the Korean Genome and Epidemiology Study (KoGES) were analyzed. The METS-IR was calculated using the following formula: ln [(2 × fasting blood glucose (FBG) + triglycerides (TG)) × body mass index (BMI)]/ ln [high-density lipoprotein cholesterol (HDL-C)]. The participants were divided into tertiles 1 (T1) and 3 (T3) based on their METS-IR scores. Genome-wide association studies (GWAS) were performed for hypertensive cases and non-hypertensive controls within these tertile groups using logistic regression adjusted for age, sex, and lifestyle factors. Among the METS-IR tertile groups, 3517 of the 19,774 participants (17.8%) at T1 had hypertension, whereas 8653 of the 20,374 participants (42.5%) at T3 had hypertension. A total of 113 single-nucleotide polymorphisms (SNPs) reached the GWAS significance threshold (p < 5 × 10-8) in at least one tertile group, mapping to six distinct genetic loci. Notably, four loci, rs11899121 (chr2p24), rs7556898 (chr2q24.3), rs17249754 (ATP2B1), and rs1980854 (chr20p12.2), were significantly associated with hypertension in the high-METS-score group (T3). rs10857147 (FGF5) was significant in both the T1 and T3 groups, whereas rs671 (ALDH2) was significant only in the T1 group. The GWASs identified six genetic loci significantly associated with hypertension, with distinct patterns across METS-IR tertiles, highlighting the role of metabolic context in genetic susceptibility. These findings underscore critical genetic factors influencing hypertension prevalence and provide insights into the metabolic-genetic interplay underlying this condition.
Collapse
Affiliation(s)
- Bo-Kyung Shine
- Department of Family Medicine, Medical Center, Dong-A University, Busan 49201, Republic of Korea
| | - Ja-Eun Choi
- Institute of Advanced Technology, Theragen Health Co., Ltd., Seongnam 13493, Republic of Korea
| | - Young-Jin Park
- Department of Family Medicine, Medical Center, Dong-A University, Busan 49201, Republic of Korea
| | - Kyung-Won Hong
- Institute of Advanced Technology, Theragen Health Co., Ltd., Seongnam 13493, Republic of Korea
| |
Collapse
|
8
|
Moore A, Ritchie MD. Is the Relationship Between Cardiovascular Disease and Alzheimer's Disease Genetic? A Scoping Review. Genes (Basel) 2024; 15:1509. [PMID: 39766777 PMCID: PMC11675426 DOI: 10.3390/genes15121509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Cardiovascular disease (CVD) and Alzheimer's disease (AD) are two diseases highly prevalent in the aging population and often co-occur. The exact relationship between the two diseases is uncertain, though epidemiological studies have demonstrated that CVDs appear to increase the risk of AD and vice versa. This scoping review aims to examine the current identified overlapping genetics between CVDs and AD at the individual gene level and at the shared pathway level. METHODS Following PRISMA-ScR guidelines for a scoping review, we searched the PubMed and Scopus databases from 1990 to October 2024 for articles that involved (1) CVDs, (2) AD, and (3) used statistical methods to parse genetic relationships. RESULTS Our search yielded 2918 articles, of which 274 articles passed screening and were organized into two main sections: (1) evidence of shared genetic risk; and (2) shared mechanisms. The genes APOE, PSEN1, and PSEN2 reportedly have wide effects across the AD and CVD spectrum, affecting both cardiac and brain tissues. Mechanistically, changes in three main pathways (lipid metabolism, blood pressure regulation, and the breakdown of the blood-brain barrier (BBB)) contribute to subclinical and etiological changes that promote both AD and CVD progression. However, genetic studies continue to be limited by the availability of longitudinal data and lack of cohorts that are representative of diverse populations. CONCLUSIONS Highly penetrant familial genes simultaneously increase the risk of CVDs and AD. However, in most cases, sets of dysregulated genes within larger-scale mechanisms, like changes in lipid metabolism, blood pressure regulation, and BBB breakdown, increase the risk of both AD and CVDs and contribute to disease progression.
Collapse
Affiliation(s)
- Anni Moore
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Marylyn D. Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Division of Informatics, Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
9
|
Fan Y, Chen J, Fan Z, Chirinos J, Stein JL, Sullivan PF, Wang R, Nadig A, Zhang DY, Huang S, Jiang Z, Guan PY, Qian X, Li T, Li H, Sun Z, Ritchie MD, O’Brien J, Witschey W, Rader DJ, Li T, Zhu H, Zhao B. Mapping rare protein-coding variants on multi-organ imaging traits. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.16.24317443. [PMID: 39606337 PMCID: PMC11601754 DOI: 10.1101/2024.11.16.24317443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Human organ structure and function are important endophenotypes for clinical outcomes. Genome-wide association studies (GWAS) have identified numerous common variants associated with phenotypes derived from magnetic resonance imaging (MRI) of the brain and body. However, the role of rare protein-coding variations affecting organ size and function is largely unknown. Here we present an exome-wide association study that evaluates 596 multi-organ MRI traits across over 50,000 individuals from the UK Biobank. We identified 107 variant-level associations and 224 gene-based burden associations (67 unique gene-trait pairs) across all MRI modalities, including PTEN with total brain volume, TTN with regional peak circumferential strain in the heart left ventricle, and TNFRSF13B with spleen volume. The singleton burden model and AlphaMissense annotations contributed 8 unique gene-trait pairs including the association between an approved drug target gene of KCNA5 and brain functional activity. The identified rare coding signals elucidate some shared genetic regulation across organs, prioritize previously identified GWAS loci, and are enriched for drug targets. Overall, we demonstrate how rare variants enhance our understanding of genetic effects on human organ morphology and function and their connections to complex diseases.
Collapse
Affiliation(s)
- Yijun Fan
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jie Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zirui Fan
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julio Chirinos
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, 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
| | - Patrick F. Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rujin Wang
- Regeneron Genetics Center, 777 Old Saw Mill River Rd., Tarrytown, NY, 10591, USA
| | - Ajay Nadig
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David Y. Zhang
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shuai Huang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zhiwen Jiang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Peter Yi Guan
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xinjie Qian
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ting Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Haoyue Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zehui Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marylyn D. Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
| | - Joan O’Brien
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Medicine Center for Ophthalmic Genetics in Complex Diseases, Philadelphia, PA 19104, USA
| | - Walter Witschey
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel J. Rader
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, 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
| | - 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
| | - Bingxin Zhao
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Population Aging Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Center for Eye-Brain Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Beeche C, Dib MJ, Zhao B, Azzo JD, Tavolinejad H, Maynard H, Duda J, Gee J, Salman O, Witschey WR, Chirinos JA. Thoracic Aortic Three-Dimensional Geometry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593413. [PMID: 38798566 PMCID: PMC11118285 DOI: 10.1101/2024.05.09.593413] [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: 05/29/2024]
Abstract
Background Aortic structure impacts cardiovascular health through multiple mechanisms. Aortic structural degeneration occurs with aging, increasing left ventricular afterload and promoting increased arterial pulsatility and target organ damage. Despite the impact of aortic structure on cardiovascular health, three-dimensional (3D) aortic geometry has not been comprehensively characterized in large populations. Methods We segmented the complete thoracic aorta using a deep learning architecture and used morphological image operations to extract multiple aortic geometric phenotypes (AGPs, including diameter, length, curvature, and tortuosity) across various subsegments of the thoracic aorta. We deployed our segmentation approach on imaging scans from 54,241 participants in the UK Biobank and 8,456 participants in the Penn Medicine Biobank. Conclusion Our method provides a fully automated approach towards quantifying the three-dimensional structural parameters of the aorta. This approach expands the available phenotypes in two large representative biobanks and will allow large-scale studies to elucidate the biology and clinical consequences of aortic degeneration related to aging and disease states.
Collapse
Affiliation(s)
- Cameron Beeche
- Department of Bioengineering, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104
| | - Marie-Joe Dib
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 3400 Spruce Street, Philadelphia, PA, 19104
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104
| | - Joe David Azzo
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 3400 Spruce Street, Philadelphia, PA, 19104
| | - Hamed Tavolinejad
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104
- Department of Statistics and Data Science, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104
| | - Hannah Maynard
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104
| | - Jeffrey Duda
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104
| | - James Gee
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104
| | - Oday Salman
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 3400 Spruce Street, Philadelphia, PA, 19104
| | | | - Walter R. Witschey
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104
| | - Julio A. Chirinos
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 3400 Spruce Street, Philadelphia, PA, 19104
| |
Collapse
|
11
|
Grilo LF, Zimmerman KD, Puppala S, Chan J, Huber HF, Li G, Jadhav AYL, Wang B, Li C, Clarke GD, Register TC, Oliveira PJ, Nathanielsz PW, Olivier M, Pereira SP, Cox LA. Cardiac Molecular Analysis Reveals Aging-Associated Metabolic Alterations Promoting Glycosaminoglycans Accumulation via Hexosamine Biosynthetic Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309211. [PMID: 39119859 PMCID: PMC11481188 DOI: 10.1002/advs.202309211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 07/17/2024] [Indexed: 08/10/2024]
Abstract
Age is a prominent risk factor for cardiometabolic disease, often leading to heart structural and functional changes. However, precise molecular mechanisms underlying cardiac remodeling and dysfunction exclusively resulting from physiological aging remain elusive. Previous research demonstrated age-related functional alterations in baboons, analogous to humans. The goal of this study is to identify early cardiac molecular alterations preceding functional adaptations, shedding light on the regulation of age-associated changes. Unbiased transcriptomics of left ventricle samples are performed from female baboons aged 7.5-22.1 years (human equivalent ≈30-88 years). Weighted-gene correlation network and pathway enrichment analyses are performed, with histological validation. Modules of transcripts negatively correlated with age implicated declined metabolism-oxidative phosphorylation, tricarboxylic acid cycle, glycolysis, and fatty-acid β-oxidation. Transcripts positively correlated with age suggested a metabolic shift toward glucose-dependent anabolic pathways, including hexosamine biosynthetic pathway (HBP). This shift is associated with increased glycosaminoglycan synthesis, modification, precursor synthesis via HBP, and extracellular matrix accumulation, verified histologically. Upregulated extracellular matrix-induced signaling coincided with glycosaminoglycan accumulation, followed by cardiac hypertrophy-related pathways. Overall, these findings revealed a transcriptional shift in metabolism favoring glycosaminoglycan accumulation through HBP before cardiac hypertrophy. Unveiling this metabolic shift provides potential targets for age-related cardiac diseases, offering novel insights into early age-related mechanisms.
Collapse
Affiliation(s)
- Luís F. Grilo
- CNC‐UCCenter for Neuroscience and Cell BiologyUniversity of CoimbraCoimbra3060Portugal
- CIBBCenter for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbra3060Portugal
- Institute for Interdisciplinary ResearchPDBEB – Doctoral Programme in Experimental Biology and BiomedicineUniversity of CoimbraCoimbra3060Portugal
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
| | - Kip D. Zimmerman
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
- Section on Molecular MedicineDepartment of Internal MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
| | - Sobha Puppala
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
- Section on Molecular MedicineDepartment of Internal MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
| | - Jeannie Chan
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
- Section on Molecular MedicineDepartment of Internal MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
| | - Hillary F. Huber
- Southwest National Primate Research CenterTexas Biomedical Research InstituteSan AntonioTX78245USA
| | - Ge Li
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
| | - Avinash Y. L. Jadhav
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
| | - Benlian Wang
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
| | - Cun Li
- Texas Pregnancy & Life‐Course Health Research CenterDepartment of Animal ScienceUniversity of WyomingLaramieWY82071USA
| | - Geoffrey D. Clarke
- Department of RadiologyUniversity of Texas Health Science CenterSan AntonioTX78229USA
| | - Thomas C. Register
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC27157USA
| | - Paulo J. Oliveira
- CNC‐UCCenter for Neuroscience and Cell BiologyUniversity of CoimbraCoimbra3060Portugal
- CIBBCenter for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbra3060Portugal
| | - Peter W. Nathanielsz
- Texas Pregnancy & Life‐Course Health Research CenterDepartment of Animal ScienceUniversity of WyomingLaramieWY82071USA
| | - Michael Olivier
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
- Section on Molecular MedicineDepartment of Internal MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
| | - Susana P. Pereira
- CNC‐UCCenter for Neuroscience and Cell BiologyUniversity of CoimbraCoimbra3060Portugal
- CIBBCenter for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbra3060Portugal
- Laboratory of Metabolism and Exercise (LaMetEx)Research Centre in Physical ActivityHealth and Leisure (CIAFEL)Laboratory for Integrative and Translational Research in Population Health (ITR)Faculty of SportsUniversity of PortoPorto4050Portugal
| | - Laura A. Cox
- Center for Precision MedicineWake Forest University Health SciencesWinston‐SalemNC27157USA
- Section on Molecular MedicineDepartment of Internal MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
- Southwest National Primate Research CenterTexas Biomedical Research InstituteSan AntonioTX78245USA
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC27157USA
| |
Collapse
|
12
|
Wang M, Wang Z, Wang Y, Zhou Q, Wang J. Causal relationships involving brain imaging-derived phenotypes based on UKB imaging cohort: a review of Mendelian randomization studies. Front Neurosci 2024; 18:1436223. [PMID: 39050670 PMCID: PMC11266110 DOI: 10.3389/fnins.2024.1436223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024] Open
Abstract
The UK Biobank (UKB) has the largest adult brain imaging dataset, which encompasses over 40,000 participants. A significant number of Mendelian randomization (MR) studies based on UKB neuroimaging data have been published to validate potential causal relationships identified in observational studies. Relevant articles published before December 2023 were identified following the PRISMA protocol. Included studies (n = 34) revealed that there were causal relationships between various lifestyles, diseases, biomarkers, and brain image-derived phenotypes (BIDPs). In terms of lifestyle habits and environmental factors, there were causal relationships between alcohol consumption, tea intake, coffee consumption, smoking, educational attainment, and certain BIDPs. Additionally, some BIDPs could serve as mediators between leisure/physical inactivity and major depressive disorder. Regarding diseases, BIDPs have been found to have causal relationships not only with Alzheimer's disease, stroke, psychiatric disorders, and migraine, but also with cardiovascular diseases, diabetes, poor oral health, osteoporosis, and ankle sprain. In addition, there were causal relationships between certain biological markers and BIDPs, such as blood pressure, LDL-C, IL-6, telomere length, and more.
Collapse
Affiliation(s)
- Mengdong Wang
- Department of Radiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zirui Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Yaoyi Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Quan Zhou
- Department of Radiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Junping Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
13
|
Han QJ, Zhu YP, Sun J, Ding XY, Wang X, Zhang QZ. PTGES2 and RNASET2 identified as novel potential biomarkers and therapeutic targets for basal cell carcinoma: insights from proteome-wide mendelian randomization, colocalization, and MR-PheWAS analyses. Front Pharmacol 2024; 15:1418560. [PMID: 39035989 PMCID: PMC11257982 DOI: 10.3389/fphar.2024.1418560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/12/2024] [Indexed: 07/23/2024] Open
Abstract
Introduction Basal cell carcinoma (BCC) is the most common skin cancer, lacking reliable biomarkers or therapeutic targets for effective treatment. Genome-wide association studies (GWAS) can aid in identifying drug targets, repurposing existing drugs, predicting clinical trial side effects, and reclassifying patients in clinical utility. Hence, the present study investigates the association between plasma proteins and skin cancer to identify effective biomarkers and therapeutic targets for BCC. Methods Proteome-wide mendelian randomization was performed using inverse-variance-weight and Wald Ratio methods, leveraging 1 Mb cis protein quantitative trait loci (cis-pQTLs) in the UK Biobank Pharma Proteomics Project (UKB-PPP) and the deCODE Health Study, to determine the causal relationship between plasma proteins and skin cancer and its subtypes in the FinnGen R10 study and the SAIGE database of Lee lab. Significant association with skin cancer and its subtypes was defined as a false discovery rate (FDR) < 0.05. pQTL to GWAS colocalization analysis was executed using a Bayesian model to evaluate five exclusive hypotheses. Strong colocalization evidence was defined as a posterior probability for shared causal variants (PP.H4) of ≥0.85. Mendelian randomization-Phenome-wide association studies (MR-PheWAS) were used to evaluate potential biomarkers and therapeutic targets for skin cancer and its subtypes within a phenome-wide human disease category. Results PTGES2, RNASET2, SF3B4, STX8, ENO2, and HS3ST3B1 (besides RNASET2, five other plasma proteins were previously unknown in expression quantitative trait loci (eQTL) and methylation quantitative trait loci (mQTL)) were significantly associated with BCC after FDR correction in the UKB-PPP and deCODE studies. Reverse MR showed no association between BCC and these proteins. PTGES2 and RNASET2 exhibited strong evidence of colocalization with BCC based on a posterior probability PP.H4 >0.92. Furthermore, MR-PheWAS analysis showed that BCC was the most significant phenotype associated with PTGES2 and RNASET2 among 2,408 phenotypes in the FinnGen R10 study. Therefore, PTGES2 and RNASET2 are highlighted as effective biomarkers and therapeutic targets for BCC within the phenome-wide human disease category. Conclusion The study identifies PTGES2 and RNASET2 plasma proteins as novel, reliable biomarkers and therapeutic targets for BCC, suggesting more effective clinical application strategies for patients.
Collapse
Affiliation(s)
- Qiu-Ju Han
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, and the Haihe Laboratory of Cell Ecosystem, Tianjin, China
| | - Yi-Pan Zhu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, and the Haihe Laboratory of Cell Ecosystem, Tianjin, China
| | - Jing Sun
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, and the Haihe Laboratory of Cell Ecosystem, Tianjin, China
| | - Xin-Yu Ding
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, and the Haihe Laboratory of Cell Ecosystem, Tianjin, China
| | - Xiuyu Wang
- Department of Neurosurgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Qiang-Zhe Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, and the Haihe Laboratory of Cell Ecosystem, Tianjin, China
| |
Collapse
|
14
|
Yang ML, Xu C, Gupte T, Hoffmann TJ, Iribarren C, Zhou X, Ganesh SK. Sex-specific genetic architecture of blood pressure. Nat Med 2024; 30:818-828. [PMID: 38459180 PMCID: PMC11797078 DOI: 10.1038/s41591-024-02858-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/05/2024] [Indexed: 03/10/2024]
Abstract
The genetic and genomic basis of sex differences in blood pressure (BP) traits remain unstudied at scale. Here, we conducted sex-stratified and combined-sex genome-wide association studies of BP traits using the UK Biobank resource, identifying 1,346 previously reported and 29 new BP trait-associated loci. Among associated loci, 412 were female-specific (Pfemale ≤ 5 × 10-8; Pmale > 5 × 10-8) and 142 were male-specific (Pmale ≤ 5 × 10-8; Pfemale > 5 × 10-8); these sex-specific loci were enriched for hormone-related transcription factors, in particular, estrogen receptor 1. Analyses of gene-by-sex interactions and sexually dimorphic effects identified four genomic regions, showing female-specific associations with diastolic BP or pulse pressure, including the chromosome 13q34-COL4A1/COL4A2 locus. Notably, female-specific pulse pressure-associated loci exhibited enriched acetylated histone H3 Lys27 modifications in arterial tissues and a female-specific association with fibromuscular dysplasia, a female-biased vascular disease; colocalization signals included Chr13q34: COL4A1/COL4A2, Chr9p21: CDKN2B-AS1 and Chr4q32.1: MAP9 regions. Sex-specific and sex-biased polygenic associations of BP traits were associated with multiple cardiovascular traits. These findings suggest potentially clinically significant and BP sex-specific pleiotropic effects on cardiovascular diseases.
Collapse
Affiliation(s)
- Min-Lee Yang
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Chang Xu
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Trisha Gupte
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Thomas J Hoffmann
- Department of Epidemiology & Biostatistics, and Institute for Human Genetics, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | - Xiang Zhou
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Santhi K Ganesh
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA.
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA.
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
15
|
Liu D, Billington CJ, Raja N, Wong ZC, Levin MD, Resch W, Alba C, Hupalo DN, Biamino E, Bedeschi MF, Digilio MC, Squeo GM, Villa R, Parrish PCR, Knutsen RH, Osgood S, Freeman JA, Dalgard CL, Merla G, Pober BR, Mervis CB, Roberts AE, Morris CA, Osborne LR, Kozel BA. Matrisome and Immune Pathways Contribute to Extreme Vascular Outcomes in Williams-Beuren Syndrome. J Am Heart Assoc 2024; 13:e031377. [PMID: 38293922 PMCID: PMC11056152 DOI: 10.1161/jaha.123.031377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/28/2023] [Indexed: 02/01/2024]
Abstract
BACKGROUND Supravalvar aortic stenosis (SVAS) is a characteristic feature of Williams-Beuren syndrome (WBS). Its severity varies: ~20% of people with Williams-Beuren syndrome have SVAS requiring surgical intervention, whereas ~35% have no appreciable SVAS. The remaining individuals have SVAS of intermediate severity. Little is known about genetic modifiers that contribute to this variability. METHODS AND RESULTS We performed genome sequencing on 473 individuals with Williams-Beuren syndrome and developed strategies for modifier discovery in this rare disease population. Approaches include extreme phenotyping and nonsynonymous variant prioritization, followed by gene set enrichment and pathway-level association tests. We next used GTEx v8 and proteomic data sets to verify expression of candidate modifiers in relevant tissues. Finally, we evaluated overlap between the genes/pathways identified here and those ascertained through larger aortic disease/trait genome-wide association studies. We show that SVAS severity in Williams-Beuren syndrome is associated with increased frequency of common and rarer variants in matrisome and immune pathways. Two implicated matrisome genes (ACAN and LTBP4) were uniquely expressed in the aorta. Many genes in the identified pathways were previously reported in genome-wide association studies for aneurysm, bicuspid aortic valve, or aortic size. CONCLUSIONS Smaller sample sizes in rare disease studies necessitate new approaches to detect modifiers. Our strategies identified variation in matrisome and immune pathways that are associated with SVAS severity. These findings suggest that, like other aortopathies, SVAS may be influenced by the balance of synthesis and degradation of matrisome proteins. Leveraging multiomic data and results from larger aorta-focused genome-wide association studies may accelerate modifier discovery for rare aortopathies like SVAS.
Collapse
Affiliation(s)
- Delong Liu
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| | - Charles J. Billington
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
- Department of PediatricsUniversity of MinnesotaMinneapolisMN
| | - Neelam Raja
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| | - Zoe C. Wong
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| | - Mark D. Levin
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| | - Wulfgang Resch
- The High Performance Computing FacilityCenter for Information Technology, National Institutes of HealthBethesdaMD
| | - Camille Alba
- Henry M Jackson Foundation for the Advancement of Military MedicineBethesdaMD
| | - Daniel N. Hupalo
- Henry M Jackson Foundation for the Advancement of Military MedicineBethesdaMD
| | | | | | | | - Gabriella Maria Squeo
- Laboratory of Regulatory and Functional GenomicsFondazione IRCCS Casa Sollievo della SofferenzaSan Giovanni Rotondo (Foggia)Italy
| | - Roberta Villa
- Fondazione IRCCS Ca Granda Ospedale Maggiore Policlinico Medical Genetic UnitMilanItaly
| | - Pheobe C. R. Parrish
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
- Department of Genome SciencesUniversity of WashingtonSeattleWA
| | - Russell H. Knutsen
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| | - Sharon Osgood
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| | - Joy A. Freeman
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| | - Clifton L. Dalgard
- Department of Anatomy, Physiology and Genetics, School of Medicinethe Uniformed Services University of the Health SciencesBethesdaMD
| | - Giuseppe Merla
- Laboratory of Regulatory and Functional GenomicsFondazione IRCCS Casa Sollievo della SofferenzaSan Giovanni Rotondo (Foggia)Italy
- Department of Molecular Medicine and Medical BiotechnologyUniversity of Naples Federico IINaplesItaly
| | - Barbara R. Pober
- Section of Genetics, Department of PediatricsMassachusetts General HospitalBostonMA
| | - Carolyn B. Mervis
- Department of Psychological and Brain SciencesUniversity of LouisvilleLouisvilleKY
| | - Amy E. Roberts
- Department of Cardiology and Division of Genetics and Genomics, Department of PediatricsBoston Children’s HospitalBostonMA
| | - Colleen A. Morris
- Department of PediatricsKirk Kerkorian School of Medicine at UNLVLas VegasNV
| | - Lucy R. Osborne
- Departments of Medicine and Molecular GeneticsUniversity of TorontoCanada
| | - Beth A. Kozel
- National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMD
| |
Collapse
|
16
|
Salih AM, Galazzo IB, Menegaz G, Altmann A. Leukocyte Telomere Length and Cardiac Structure and Function: A Mendelian Randomization Study. J Am Heart Assoc 2024; 13:e032708. [PMID: 38293941 PMCID: PMC11056120 DOI: 10.1161/jaha.123.032708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/04/2024] [Indexed: 02/01/2024]
Abstract
BACKGROUND Existing research demonstrates the association of shorter leukocyte telomere length with increased risk of age-related health outcomes including cardiovascular diseases. However, the direct causality of these relationships has not been definitively established. Cardiovascular aging at an organ level may be captured using image-derived phenotypes of cardiac anatomy and function. METHODS AND RESULTS In the current study, we use 2-sample Mendelian randomization to assess the causal link between leukocyte telomere length and 54 cardiac magnetic resonance imaging measures representing structure and function across the 4 cardiac chambers. Genetically predicted shorter leukocyte telomere length was causally linked to smaller ventricular cavity sizes including left ventricular end-systolic volume, left ventricular end-diastolic volume, lower left ventricular mass, and pulmonary artery. The association with left ventricular mass (β =0.217, Pfalse discovery rate=0.016) remained significant after multiple testing adjustment, whereas other associations were attenuated. CONCLUSIONS Our findings support a causal role for shorter leukocyte telomere length and faster cardiac aging, with the most prominent relationship with left ventricular mass.
Collapse
Affiliation(s)
- Ahmed M. Salih
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of LondonUK
- Department of Population Health SciencesUniversity of LeicesterUK
- Department of Computer ScienceUniversity of ZakhoKurdistan of IraqIraq
| | | | | | - André Altmann
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical EngineeringUniversity College LondonUK
| |
Collapse
|
17
|
Munroe PB, Aung N, Ramírez J. Genetic analysis of cardiac dynamic flow volumes identifies loci mapping aortic root size. Nat Genet 2024; 56:197-199. [PMID: 38332371 DOI: 10.1038/s41588-023-01650-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Affiliation(s)
- Patricia B Munroe
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Nay Aung
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Julia Ramírez
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
- Aragon Institute of Engineering Research, University of Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina, Zaragoza, Spain
| |
Collapse
|
18
|
Gomes B, Singh A, O'Sullivan JW, Schnurr TM, Goddard PC, Loong S, Amar D, Hughes JW, Kostur M, Haddad F, Salerno M, Foo R, Montgomery SB, Parikh VN, Meder B, Ashley EA. Genetic architecture of cardiac dynamic flow volumes. Nat Genet 2024; 56:245-257. [PMID: 38082205 DOI: 10.1038/s41588-023-01587-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 10/23/2023] [Indexed: 02/04/2024]
Abstract
Cardiac blood flow is a critical determinant of human health. However, the definition of its genetic architecture is limited by the technical challenge of capturing dynamic flow volumes from cardiac imaging at scale. We present DeepFlow, a deep-learning system to extract cardiac flow and volumes from phase-contrast cardiac magnetic resonance imaging. A mixed-linear model applied to 37,653 individuals from the UK Biobank reveals genome-wide significant associations across cardiac dynamic flow volumes spanning from aortic forward velocity to aortic regurgitation fraction. Mendelian randomization reveals a causal role for aortic root size in aortic valve regurgitation. Among the most significant contributing variants, localizing genes (near ELN, PRDM6 and ADAMTS7) are implicated in connective tissue and blood pressure pathways. Here we show that DeepFlow cardiac flow phenotyping at scale, combined with genotyping data, reinforces the contribution of connective tissue genes, blood pressure and root size to aortic valve function.
Collapse
Affiliation(s)
- Bruna Gomes
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Cardiology, Pneumology and Angiology, Heidelberg University Hospital, Heidelberg, Germany
- Informatics for Life, Heidelberg, Germany
| | - Aditya Singh
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Jack W O'Sullivan
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Theresia M Schnurr
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Pagé C Goddard
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Shaun Loong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Amar
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - J Weston Hughes
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Mykhailo Kostur
- Department of Cardiology, Pneumology and Angiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Francois Haddad
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Michael Salerno
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Roger Foo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Stephen B Montgomery
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Victoria N Parikh
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Benjamin Meder
- Department of Cardiology, Pneumology and Angiology, Heidelberg University Hospital, Heidelberg, Germany
- Informatics for Life, Heidelberg, Germany
| | - Euan A Ashley
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA.
| |
Collapse
|
19
|
Raghavan A, Pirruccello JP, Ellinor PT, Lindsay ME. Using Genomics to Identify Novel Therapeutic Targets for Aortic Disease. Arterioscler Thromb Vasc Biol 2024; 44:334-351. [PMID: 38095107 PMCID: PMC10843699 DOI: 10.1161/atvbaha.123.318771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 11/21/2023] [Indexed: 01/04/2024]
Abstract
Aortic disease, including dissection, aneurysm, and rupture, carries significant morbidity and mortality and is a notable cause of sudden cardiac death. Much of our knowledge regarding the genetic basis of aortic disease has relied on the study of individuals with Mendelian aortopathies and, until recently, the genetic determinants of population-level variance in aortic phenotypes remained unclear. However, the application of machine learning methodologies to large imaging datasets has enabled researchers to rapidly define aortic traits and mine dozens of novel genetic associations for phenotypes such as aortic diameter and distensibility. In this review, we highlight the emerging potential of genomics for identifying causal genes and candidate drug targets for aortic disease. We describe how deep learning technologies have accelerated the pace of genetic discovery in this field. We then provide a blueprint for translating genetic associations to biological insights, reviewing techniques for locus and cell type prioritization, high-throughput functional screening, and disease modeling using cellular and animal models of aortic disease.
Collapse
Affiliation(s)
- Avanthi Raghavan
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - James P. Pirruccello
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
| | - Patrick T. Ellinor
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Mark E. Lindsay
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Bohoran TA, Parke KS, Graham-Brown MPM, Meisuria M, Singh A, Wormleighton J, Adlam D, Gopalan D, Davies MJ, Williams B, Brown M, McCann GP, Giannakidis A. Resource efficient aortic distensibility calculation by end to end spatiotemporal learning of aortic lumen from multicentre multivendor multidisease CMR images. Sci Rep 2023; 13:21794. [PMID: 38066222 PMCID: PMC10709583 DOI: 10.1038/s41598-023-48986-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Aortic distensibility (AD) is important for the prognosis of multiple cardiovascular diseases. We propose a novel resource-efficient deep learning (DL) model, inspired by the bi-directional ConvLSTM U-Net with densely connected convolutions, to perform end-to-end hierarchical learning of the aorta from cine cardiovascular MRI towards streamlining AD quantification. Unlike current DL aortic segmentation approaches, our pipeline: (i) performs simultaneous spatio-temporal learning of the video input, (ii) combines the feature maps from the encoder and decoder using non-linear functions, and (iii) takes into account the high class imbalance. By using multi-centre multi-vendor data from a highly heterogeneous patient cohort, we demonstrate that the proposed method outperforms the state-of-the-art method in terms of accuracy and at the same time it consumes [Formula: see text] 3.9 times less fuel and generates [Formula: see text] 2.8 less carbon emissions. Our model could provide a valuable tool for exploring genome-wide associations of the AD with the cognitive performance in large-scale biomedical databases. By making energy usage and carbon emissions explicit, the presented work aligns with efforts to keep DL's energy requirements and carbon cost in check. The improved resource efficiency of our pipeline might open up the more systematic DL-powered evaluation of the MRI-derived aortic stiffness.
Collapse
Affiliation(s)
- Tuan Aqeel Bohoran
- School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Kelly S Parke
- Department of Cardiovascular Sciences, University of Leicester and the NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Matthew P M Graham-Brown
- Department of Cardiovascular Sciences, University of Leicester and the NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Mitul Meisuria
- Department of Cardiovascular Sciences, University of Leicester and the NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Anvesha Singh
- Department of Cardiovascular Sciences, University of Leicester and the NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Joanne Wormleighton
- Department of Cardiovascular Sciences, University of Leicester and the NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - David Adlam
- Department of Cardiovascular Sciences, University of Leicester and the NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Deepa Gopalan
- Imperial College London & Cambridge University Hospitals, Cambridge, CB2 0QQ, UK
| | - Melanie J Davies
- Leicester Diabetes Centre, University of Leicester and the NIHR Leicester Biomedical Research Centre, Leicester General Hospital, Leicester, LE5 4PW, UK
| | - Bryan Williams
- Institute of Cardiovascular Science, University College London (UCL), National Institute for Health Research (NIHR), UCL Hospitals Biomedical Research Centre, London, WC1E 6DD, UK
| | - Morris Brown
- Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Gerry P McCann
- Department of Cardiovascular Sciences, University of Leicester and the NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Archontis Giannakidis
- School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK.
| |
Collapse
|
21
|
Li Z, Xiong J, Guo Y, Tang H, Guo B, Wang B, Gao D, Dong Z, Tu Y. Effects of diabetes mellitus and glycemic traits on cardiovascular morpho-functional phenotypes. Cardiovasc Diabetol 2023; 22:336. [PMID: 38066511 PMCID: PMC10709859 DOI: 10.1186/s12933-023-02079-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND The effects of diabetes on the cardiac and aortic structure and function remain unclear. Detecting and intervening these variations early is crucial for the prevention and management of complications. Cardiovascular magnetic resonance imaging-derived traits are established endophenotypes and serve as precise, early-detection, noninvasive clinical risk biomarkers. We conducted a Mendelian randomization (MR) study to examine the association between two types of diabetes, four glycemic traits, and preclinical endophenotypes of cardiac and aortic structure and function. METHODS Independent genetic variants significantly associated with type 1 diabetes, type 2 diabetes, fasting insulin (FIns), fasting glucose (FGlu), 2 h-glucose post-challenge (2hGlu), and glycated hemoglobin (HbA1c) were selected as instrumental variables. The 96 cardiovascular magnetic resonance imaging traits came from six independent genome-wide association studies. These traits serve as preclinical endophenotypes and offer an early indication of the structure and function of the four cardiac chambers and two aortic sections. The primary analysis was performed using MR with the inverse-variance weighted method. Confirmation was achieved through Steiger filtering and testing to determine the causal direction. Sensitivity analyses were conducted using the weighted median, MR-Egger, and MR-PRESSO methods. Additionally, multivariable MR was used to adjust for potential effects associated with body mass index. RESULTS Genetic susceptibility to type 1 diabetes was associated with increased ascending aortic distensibility. Conversely, type 2 diabetes showed a correlation with a reduced diameter and areas of the ascending aorta, as well as decreased distensibility of the descending aorta. Genetically predicted higher levels of FGlu and HbA1c were correlated with a decrease in diameter and areas of the ascending aorta. Furthermore, higher 2hGlu levels predominantly showed association with a reduced diameter of both the ascending and descending aorta. Higher FIns levels corresponded to increased regional myocardial-wall thicknesses at end-diastole, global myocardial-wall thickness at end-diastole, and regional peak circumferential strain of the left ventricle. CONCLUSIONS This study provides evidence that diabetes and glycemic traits have a causal relationship with cardiac and aortic structural and functional remodeling, highlighting the importance of intensive glucose-lowering for primary prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Zhaoyue Li
- Harbin Medical University, Harbin, China
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jie Xiong
- Harbin Medical University, Harbin, China
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yutong Guo
- Harbin Medical University, Harbin, China
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Hao Tang
- Harbin Medical University, Harbin, China
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Bingchen Guo
- Harbin Medical University, Harbin, China
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Bo Wang
- Harbin Medical University, Harbin, China
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Dianyu Gao
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zengxiang Dong
- Harbin Medical University, Harbin, China.
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Yingfeng Tu
- Harbin Medical University, Harbin, China.
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin, China.
| |
Collapse
|
22
|
Grilo LF, Zimmerman KD, Puppala S, Chan J, Huber HF, Li G, Jadhav AYL, Wang B, Li C, Clarke GD, Register TC, Oliveira PJ, Nathanielsz PW, Olivier M, Pereira SP, Cox LA. Cardiac Molecular Analysis Reveals Aging-Associated Metabolic Alterations Promoting Glycosaminoglycans Accumulation Via Hexosamine Biosynthetic Pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.567640. [PMID: 38014295 PMCID: PMC10680868 DOI: 10.1101/2023.11.17.567640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Age is a prominent risk factor for cardiometabolic disease, and often leads to heart structural and functional changes. However, precise molecular mechanisms underlying cardiac remodeling and dysfunction resulting from physiological aging per se remain elusive. Understanding these mechanisms requires biological models with optimal translation to humans. Previous research demonstrated that baboons undergo age-related reduction in ejection fraction and increased heart sphericity, mirroring changes observed in humans. The goal of this study was to identify early cardiac molecular alterations that precede functional adaptations, shedding light on the regulation of age-associated changes. We performed unbiased transcriptomics of left ventricle (LV) samples from female baboons aged 7.5-22.1 years (human equivalent ~30-88 years). Weighted-gene correlation network and pathway enrichment analyses were performed to identify potential age-associated mechanisms in LV, with histological validation. Myocardial modules of transcripts negatively associated with age were primarily enriched for cardiac metabolism, including oxidative phosphorylation, tricarboxylic acid cycle, glycolysis, and fatty-acid β-oxidation. Transcripts positively correlated with age suggest upregulation of glucose uptake, pentose phosphate pathway, and hexosamine biosynthetic pathway (HBP), indicating a metabolic shift towards glucose-dependent anabolic pathways. Upregulation of HBP commonly results in increased glycosaminoglycan precursor synthesis. Transcripts involved in glycosaminoglycan synthesis, modification, and intermediate metabolism were also upregulated in older animals, while glycosaminoglycan degradation transcripts were downregulated with age. These alterations would promote glycosaminoglycan accumulation, which was verified histologically. Upregulation of extracellular matrix (ECM)-induced signaling pathways temporally coincided with glycosaminoglycan accumulation. We found a subsequent upregulation of cardiac hypertrophy-related pathways and an increase in cardiomyocyte width. Overall, our findings revealed a transcriptional shift in metabolism from catabolic to anabolic pathways that leads to ECM glycosaminoglycan accumulation through HBP prior to upregulation of transcripts of cardiac hypertrophy-related pathways. This study illuminates cellular mechanisms that precede development of cardiac hypertrophy, providing novel potential targets to remediate age-related cardiac diseases.
Collapse
Affiliation(s)
- Luís F. Grilo
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
- University of Coimbra, Institute for Interdisciplinary Research, PDBEB - Doctoral Programme in Experimental Biology and Biomedicine
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Kip D. Zimmerman
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Sobha Puppala
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Jeannie Chan
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Hillary F. Huber
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Ge Li
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Avinash Y. L. Jadhav
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Benlian Wang
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Cun Li
- Texas Pregnancy & Life-Course Health Research Center, Department of Animal Science, University of Wyoming, Laramie, Wyoming, USA
| | - Geoffrey D. Clarke
- Department of Radiology, University of Texas Health Science Center, San Antonio, Texas
| | - Thomas C. Register
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
- Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Paulo J. Oliveira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
| | - Peter W. Nathanielsz
- Texas Pregnancy & Life-Course Health Research Center, Department of Animal Science, University of Wyoming, Laramie, Wyoming, USA
| | - Michael Olivier
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Susana P. Pereira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| | - Laura A. Cox
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
- Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
23
|
Le Grand Q, Ecker Ferreira L, Metso TM, Schilling S, Tatlisumak T, Grond-Ginsbach C, Engelter ST, Lyrer P, Majersik JJ, Worrall BB, Southerland AM, Markus HS, Lathrop M, Thijs V, Leys D, Amouyel P, Dallongeville J, Dichgans M, Pezzini A, Bersano A, Sargurupremraj M, Debette S. Genetic Insights on the Relation of Vascular Risk Factors and Cervical Artery Dissection. J Am Coll Cardiol 2023; 82:1411-1423. [PMID: 37758436 DOI: 10.1016/j.jacc.2023.07.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/16/2023] [Accepted: 07/24/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND The association between vascular risk factors and cervical artery dissections (CeADs), a leading cause of ischemic stroke (IS) in the young, remains controversial. OBJECTIVES This study aimed to explore the causal relation of vascular risk factors with CeAD risk and recurrence and compare it to their relation with non-CeAD IS. METHODS This study used 2-sample Mendelian randomization analyses to explore the association of blood pressure (BP), lipid levels, type 2 diabetes, waist-to-hip ratio, smoking, and body mass index with CeAD and non-CeAD IS. To simulate effects of the most frequently used BP-lowering drugs, this study constructed genetic proxies and tested their association with CeAD and non-CeAD IS. In analyses among patients with CeAD, the investigators studied the association between weighted genetic risk scores of vascular risk factors and the risk of multiple or early recurrent dissections. RESULTS Genetically determined higher systolic BP (OR: 1.51; 95% CI: 1.32-1.72) and diastolic BP (OR: 2.40; 95% CI: 1.92-3.00) increased the risk of CeAD (P < 0.0001). Genetically determined higher body mass index was inconsistently associated with a lower risk of CeAD. Genetic proxies for β-blocker effects were associated with a lower risk of CeAD (OR: 0.65; 95% CI: 0.50-0.85), whereas calcium-channel blockers were associated with a lower risk of non-CeAD IS (OR: 0.75; 95% CI: 0.63-0.90). Weighted genetic risk scores for systolic BP and diastolic BP were associated with an increased risk of multiple or early recurrent CeAD. CONCLUSIONS These results are supportive of a causal association between higher BP and increased CeAD risk and recurrence and provide genetic evidence for lower CeAD risk under β-blockers. This may inform secondary prevention strategies and trial design for CeAD.
Collapse
Affiliation(s)
- Quentin Le Grand
- University of Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM), Bordeaux Population Health Center (BPH), Unité Mixte de Recherche (U) 1219, Bordeaux, France
| | - Leslie Ecker Ferreira
- Department of Medicine and Joinville Stroke Biobank, University of Region of Joinville, Joinville, Brazil
| | - Tiina M Metso
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
| | - Sabrina Schilling
- University of Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM), Bordeaux Population Health Center (BPH), Unité Mixte de Recherche (U) 1219, Bordeaux, France
| | - Turgut Tatlisumak
- Department of Clinical Neurosciences/Neurology, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden; Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Caspar Grond-Ginsbach
- Department of Vascular and Endovascular Surgery, University of Heidelberg, Heidelberg, Germany
| | - Stefan T Engelter
- Neurology and Neurorehabilitation, University Department of Geriatric Medicine Felix Platter, University of Basel, Basel, Switzerland; Department of Neurology and Stroke Center, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Philippe Lyrer
- Department of Neurology and Stroke Center, University Hospital Basel, University of Basel, Basel, Switzerland
| | | | - Bradford B Worrall
- Department of Neurology, University of Virginia, Charlottesville, Virginia, USA; Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Andrew M Southerland
- Department of Neurology, University of Virginia, Charlottesville, Virginia, USA; Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Hugh S Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Mark Lathrop
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada; Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Quebec, Canada
| | - Vincent Thijs
- Stroke Division, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia; Department of Neurology, Austin Health, Melbourne, Victoria, Australia
| | - Didier Leys
- INSERM U1172, Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - Philippe Amouyel
- Laboratory of Excellence Development of Innovative Strategies for a Transdisciplinary Approach to Alzheimer's Disease (LabEx DISTALZ), University of Lille, Lille, France; INSERM U1167 (Risk Factors and Molecular Determinants of Aging-Related Diseases - RID-AGE), Lille, France; Centre Hospitalier Universitaire Lille, Lille, France; Institut Pasteur de Lille, Lille, France
| | - Jean Dallongeville
- Laboratory of Excellence Development of Innovative Strategies for a Transdisciplinary Approach to Alzheimer's Disease (LabEx DISTALZ), University of Lille, Lille, France; INSERM U1167 (Risk Factors and Molecular Determinants of Aging-Related Diseases - RID-AGE), Lille, France; Centre Hospitalier Universitaire Lille, Lille, France; Institut Pasteur de Lille, Lille, France
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Munich Cluster for Systems Neurology, Munich, Germany; German Center for Neurodegenerative Diseases, Munich, Germany
| | - Alessandro Pezzini
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
| | - Anna Bersano
- Cerebrovascular Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico Carlo Besta, Milan, Italy
| | - Muralidharan Sargurupremraj
- University of Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM), Bordeaux Population Health Center (BPH), Unité Mixte de Recherche (U) 1219, Bordeaux, France; Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, Texas
| | - Stéphanie Debette
- University of Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM), Bordeaux Population Health Center (BPH), Unité Mixte de Recherche (U) 1219, Bordeaux, France; Department of Neurology, Institute of Neurodegenerative Diseases, Bordeaux University Hospital, Bordeaux, France.
| |
Collapse
|
24
|
Lum RTW, Wang X, Zhang M, Zhang X, Ho JYK, Chow SCY, Fujikawa T, Wong RHL. Emerging role of radiogenomics in genetically triggered thoracic aortic aneurysm and dissection: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:356. [PMID: 37675315 PMCID: PMC10477616 DOI: 10.21037/atm-22-6149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 03/17/2023] [Indexed: 09/08/2023]
Abstract
Background and Objective Thoracic aortic aneurysm and dissection (TAAD) and its complications are life-threatening conditions. Hypertension and atherosclerosis had all along been recognized as the predominant risk factors for the development of TAAD. However, it was increasingly reported that genetic factors, such as single nucleotide polymorphisms (SNPs), are playing an important role in the disease development. The development of next-generation sequencing (NGS) and the rapid growth in radiomics provide a promising new platform to evaluate genetically triggered thoracic aortic aneurysm and dissection (GTAAD) from a new angle. This review is to present an overview of currently available knowledge regarding the use of radiomics and radiogenomics in GTAAD. Methods We performed literature searches in PubMed, EMBASE and Cochrane database from 2012 to 2022 regarding the use of radiomics and radiogenomics in GTAAD. Key Content and Findings There were only 13 studies on radiomics and 4 studies on radiogenomics integration retrieved from the search and it signifies there is still a significant knowledge gap in this field of translational medicine. An overview of the current knowledge of GTAAD, the workflow and role of radiomics, the radiogenomics integration for GTAAD including its potential role in the development of polygenic scores, as well as the implications, challenges, and limitations of radiogenomics research were discussed. Conclusions In the contemporary era, radiogenomics has been emerging as a state-of-the art approach to establish statistical correlation with radiomics features with genomic information in diagnosis, risk modeling and prediction and treatment decision in TAAD.
Collapse
Affiliation(s)
- Ray Tak Wai Lum
- Division of Cardiothoracic Surgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Xin Wang
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Miaoru Zhang
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Xianrui Zhang
- Department of Biomedical Science, The City University of Hong Kong, Hong Kong, China
| | - Jacky Yan Kit Ho
- Division of Cardiothoracic Surgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Simon Chi Ying Chow
- Division of Cardiothoracic Surgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Takuya Fujikawa
- Division of Cardiothoracic Surgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Randolph Hung Leung Wong
- Division of Cardiothoracic Surgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
25
|
Shah M, de A Inácio MH, Lu C, Schiratti PR, Zheng SL, Clement A, de Marvao A, Bai W, King AP, Ware JS, Wilkins MR, Mielke J, Elci E, Kryukov I, McGurk KA, Bender C, Freitag DF, O'Regan DP. Environmental and genetic predictors of human cardiovascular ageing. Nat Commun 2023; 14:4941. [PMID: 37604819 PMCID: PMC10442405 DOI: 10.1038/s41467-023-40566-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 08/02/2023] [Indexed: 08/23/2023] Open
Abstract
Cardiovascular ageing is a process that begins early in life and leads to a progressive change in structure and decline in function due to accumulated damage across diverse cell types, tissues and organs contributing to multi-morbidity. Damaging biophysical, metabolic and immunological factors exceed endogenous repair mechanisms resulting in a pro-fibrotic state, cellular senescence and end-organ damage, however the genetic architecture of cardiovascular ageing is not known. Here we use machine learning approaches to quantify cardiovascular age from image-derived traits of vascular function, cardiac motion and myocardial fibrosis, as well as conduction traits from electrocardiograms, in 39,559 participants of UK Biobank. Cardiovascular ageing is found to be significantly associated with common or rare variants in genes regulating sarcomere homeostasis, myocardial immunomodulation, and tissue responses to biophysical stress. Ageing is accelerated by cardiometabolic risk factors and we also identify prescribed medications that are potential modifiers of ageing. Through large-scale modelling of ageing across multiple traits our results reveal insights into the mechanisms driving premature cardiovascular ageing and reveal potential molecular targets to attenuate age-related processes.
Collapse
Affiliation(s)
- Mit Shah
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Marco H de A Inácio
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Chang Lu
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | | | - Sean L Zheng
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Adam Clement
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Antonio de Marvao
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Wenjia Bai
- Department of Computing, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Andrew P King
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - James S Ware
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Johanna Mielke
- Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany
| | - Eren Elci
- Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany
| | - Ivan Kryukov
- Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany
| | - Kathryn A McGurk
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Christian Bender
- Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany
| | - Daniel F Freitag
- Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany
| | - Declan P O'Regan
- MRC London Institute of Medical Sciences, Imperial College London, London, UK.
| |
Collapse
|
26
|
Belmont JW. Genetic Epidemiology Highlights the Role of Aortic Strain and Distensibility in Cardiovascular Disease. J Am Coll Cardiol 2023; 81:1336-1338. [PMID: 37019579 DOI: 10.1016/j.jacc.2023.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/07/2023] [Accepted: 02/16/2023] [Indexed: 04/07/2023]
Affiliation(s)
- John W Belmont
- Departments of Molecular and Human Genetics and Pediatrics, Baylor College of Medicine, Houston, Texas, USA; and the Genetics and Genomics Services Inc, Houston, Texas, USA.
| |
Collapse
|
27
|
Chou E, Pirruccello JP, Ellinor PT, Lindsay ME. Genetics and mechanisms of thoracic aortic disease. Nat Rev Cardiol 2023; 20:168-180. [PMID: 36131050 DOI: 10.1038/s41569-022-00763-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/03/2022] [Indexed: 11/09/2022]
Abstract
Aortic disease has many forms including aortic aneurysm and dissection, aortic coarctation or abnormalities in aortic function, such as loss of aortic distensibility. Genetic analysis in humans is one of the most important experimental approaches in uncovering disease mechanisms, but the relative infrequency of thoracic aortic disease compared with other cardiovascular conditions such as coronary artery disease has hindered large-scale identification of genetic associations. In the past decade, advances in machine learning technology coupled with large imaging datasets from biobank repositories have facilitated a rapid expansion in our capacity to measure and genotype aortic traits, resulting in the identification of dozens of genetic associations. In this Review, we describe the history of technological advances in genetic discovery and explain how newer technologies such as deep learning can rapidly define aortic traits at scale. Furthermore, we integrate novel genetic observations provided by these advances into our current biological understanding of thoracic aortic disease and describe how these new findings can contribute to strategies to prevent and treat aortic disease.
Collapse
Affiliation(s)
- Elizabeth Chou
- Division of Vascular and Endovascular Surgery, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA
| | - James P Pirruccello
- Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick T Ellinor
- Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Mark E Lindsay
- Harvard Medical School, Boston, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA.
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
28
|
Chou EL, Chaffin M, Simonson B, Pirruccello JP, Akkad AD, Nekoui M, Cardenas CLL, Bedi KC, Nash C, Juric D, Stone JR, Isselbacher EM, Margulies KB, Klattenhoff C, Ellinor PT, Lindsay ME. Aortic Cellular Diversity and Quantitative Genome-Wide Association Study Trait Prioritization Through Single-Nuclear RNA Sequencing of the Aneurysmal Human Aorta. Arterioscler Thromb Vasc Biol 2022; 42:1355-1374. [PMID: 36172868 PMCID: PMC9613617 DOI: 10.1161/atvbaha.122.317953] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 09/16/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Mural cells in ascending aortic aneurysms undergo phenotypic changes that promote extracellular matrix destruction and structural weakening. To explore this biology, we analyzed the transcriptional features of thoracic aortic tissue. METHODS Single-nuclear RNA sequencing was performed on 13 samples from human donors, 6 with thoracic aortic aneurysm, and 7 without aneurysm. Individual transcriptomes were then clustered based on transcriptional profiles. Clusters were used for between-disease differential gene expression analyses, subcluster analysis, and analyzed for intersection with genetic aortic trait data. RESULTS We sequenced 71 689 nuclei from human thoracic aortas and identified 14 clusters, aligning with 11 cell types, predominantly vascular smooth muscle cells (VSMCs) consistent with aortic histology. With unbiased methodology, we found 7 vascular smooth muscle cell and 6 fibroblast subclusters. Differentially expressed genes analysis revealed a vascular smooth muscle cell group accounting for the majority of differential gene expression. Fibroblast populations in aneurysm exhibit distinct behavior with almost complete disappearance of quiescent fibroblasts. Differentially expressed genes were used to prioritize genes at aortic diameter and distensibility genome-wide association study loci highlighting the genes JUN, LTBP4 (latent transforming growth factor beta-binding protein 1), and IL34 (interleukin 34) in fibroblasts, ENTPD1, PDLIM5 (PDZ and LIM domain 5), ACTN4 (alpha-actinin-4), and GLRX in vascular smooth muscle cells, as well as LRP1 in macrophage populations. CONCLUSIONS Using nuclear RNA sequencing, we describe the cellular diversity of healthy and aneurysmal human ascending aorta. Sporadic aortic aneurysm is characterized by differential gene expression within known cellular classes rather than by the appearance of novel cellular forms. Single-nuclear RNA sequencing of aortic tissue can be used to prioritize genes at aortic trait loci.
Collapse
Affiliation(s)
- Elizabeth L. Chou
- Division of Vascular and Endovascular Surgery,
Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General
Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute,
Cambridge, Massachusetts, USA
| | - Mark Chaffin
- Cardiovascular Disease Initiative, Broad Institute,
Cambridge, Massachusetts, USA
- Precision Cardiology Laboratory, The Broad Institute,
Cambridge, MA, USA 02142
| | - Bridget Simonson
- Cardiovascular Disease Initiative, Broad Institute,
Cambridge, Massachusetts, USA
- Precision Cardiology Laboratory, The Broad Institute,
Cambridge, MA, USA 02142
| | - James P. Pirruccello
- Cardiology Division, Massachusetts General Hospital,
Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General
Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute,
Cambridge, Massachusetts, USA
- Precision Cardiology Laboratory, The Broad Institute,
Cambridge, MA, USA 02142
- Demoulas Center for Cardiac Arrhythmias, Massachusetts
General Hospital, Boston, Massachusetts, USA
| | - Amer-Denis Akkad
- Precision Cardiology Laboratory, Bayer US LLC, Cambridge,
MA, USA 02142
| | - Mahan Nekoui
- Cardiovascular Disease Initiative, Broad Institute,
Cambridge, Massachusetts, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts
General Hospital, Boston, Massachusetts, USA
| | - Christian Lacks Lino Cardenas
- Cardiology Division, Massachusetts General Hospital,
Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General
Hospital, Boston, Massachusetts, USA
| | - Kenneth C. Bedi
- Perelman School of Medicine, University of Pennsylvania,
Philadelphia, PA, USA 19104
| | - Craig Nash
- Cardiovascular Disease Initiative, Broad Institute,
Cambridge, Massachusetts, USA
- Precision Cardiology Laboratory, The Broad Institute,
Cambridge, MA, USA 02142
| | - Dejan Juric
- Cancer Center, Massachusetts General Hospital, Boston,
Massachusetts, USA
| | - James R. Stone
- Department of Pathology, Massachusetts General
Hospital, Boston, Massachusetts, USA
| | - Eric M. Isselbacher
- Cardiology Division, Massachusetts General Hospital,
Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General
Hospital, Boston, Massachusetts, USA
- Thoracic Aortic Center, Massachusetts General Hospital,
Boston, Massachusetts, USA
| | - Kenneth B. Margulies
- Perelman School of Medicine, University of Pennsylvania,
Philadelphia, PA, USA 19104
| | - Carla Klattenhoff
- Precision Cardiology Laboratory, Bayer US LLC, Cambridge,
MA, USA 02142
| | - Patrick T. Ellinor
- Cardiology Division, Massachusetts General Hospital,
Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General
Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute,
Cambridge, Massachusetts, USA
- Precision Cardiology Laboratory, The Broad Institute,
Cambridge, MA, USA 02142
- Demoulas Center for Cardiac Arrhythmias, Massachusetts
General Hospital, Boston, Massachusetts, USA
| | - Mark E. Lindsay
- Cardiology Division, Massachusetts General Hospital,
Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General
Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute,
Cambridge, Massachusetts, USA
- Thoracic Aortic Center, Massachusetts General Hospital,
Boston, Massachusetts, USA
| |
Collapse
|