1
|
Kamineni M, Raghu V, Truong B, Alaa A, Schuermans A, Friedman S, Reeder C, Bhattacharya R, Libby P, Ellinor PT, Maddah M, Philippakis A, Hornsby W, Yu Z, Natarajan P. Deep learning-derived splenic radiomics, genomics, and coronary artery disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.16.24312129. [PMID: 39185532 PMCID: PMC11343250 DOI: 10.1101/2024.08.16.24312129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Background Despite advances in managing traditional risk factors, coronary artery disease (CAD) remains the leading cause of mortality. Circulating hematopoietic cells influence risk for CAD, but the role of a key regulating organ, spleen, is unknown. The understudied spleen is a 3-dimensional structure of the hematopoietic system optimally suited for unbiased radiologic investigations toward novel mechanistic insights. Methods Deep learning-based image segmentation and radiomics techniques were utilized to extract splenic radiomic features from abdominal MRIs of 42,059 UK Biobank participants. Regression analysis was used to identify splenic radiomics features associated with CAD. Genome-wide association analyses were applied to identify loci associated with these radiomics features. Overlap between loci associated with CAD and the splenic radiomics features was explored to understand the underlying genetic mechanisms of the role of the spleen in CAD. Results We extracted 107 splenic radiomics features from abdominal MRIs, and of these, 10 features were associated with CAD. Genome-wide association analysis of CAD-associated features identified 219 loci, including 35 previously reported CAD loci, 7 of which were not associated with conventional CAD risk factors. Notably, variants at 9p21 were associated with splenic features such as run length non-uniformity. Conclusions Our study, combining deep learning with genomics, presents a new framework to uncover the splenic axis of CAD. Notably, our study provides evidence for the underlying genetic connection between the spleen as a candidate causal tissue-type and CAD with insight into the mechanisms of 9p21, whose mechanism is still elusive despite its initial discovery in 2007. More broadly, our study provides a unique application of deep learning radiomics to non-invasively find associations between imaging, genetics, and clinical outcomes.
Collapse
Affiliation(s)
| | - Vineet Raghu
- Cardiovascular Imaging Research Center, Department of Radiology, MGH and HMS
- Artificial Intelligence in Medicine Program, Mass General Brigham, Harvard Medical School, Boston, Massachusetts
| | - Buu Truong
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
| | - Ahmed Alaa
- Computational Precision Health Program, University of California, Berkeley, Berkeley, CA 94720
- Computational Precision Health Program, University of California, San Francisco, San Francisco, CA 94143
| | - Art Schuermans
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Sam Friedman
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Christopher Reeder
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Romit Bhattacharya
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston MA 02114
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115
| | - Patrick T. Ellinor
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Mahnaz Maddah
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | | | - Whitney Hornsby
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
| | - Zhi Yu
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
| | - Pradeep Natarajan
- Harvard Medical School, Boston, MA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Personalized Medicine, Mass General Brigham, Boston, MA
| |
Collapse
|
2
|
Minvielle Moncla LH, Briend M, Sokhna Sylla M, Mathieu S, Rufiange A, Bossé Y, Mathieu P. Mendelian randomization reveals interactions of the blood proteome and immunome in mitral valve prolapse. COMMUNICATIONS MEDICINE 2024; 4:108. [PMID: 38844506 PMCID: PMC11156961 DOI: 10.1038/s43856-024-00530-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/21/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Mitral valve prolapse (MVP) is a common heart disorder characterized by an excessive production of proteoglycans and extracellular matrix in mitral valve leaflets. Large-scale genome-wide association study (GWAS) underlined that MVP is heritable. The molecular underpinnings of the disease remain largely unknown. METHODS We interrogated cross-modality data totaling more than 500,000 subjects including GWAS, 4809 molecules of the blood proteome, and genome-wide expression of mitral valves to identify candidate drivers of MVP. Data were investigated through Mendelian randomization, network analysis, ligand-receptor inference and digital cell quantification. RESULTS In this study, Mendelian randomization identify that 33 blood proteins, enriched in networks for immunity, are associated with the risk of MVP. MVP- associated blood proteins are enriched in ligands for which their cognate receptors are differentially expressed in mitral valve leaflets during MVP and enriched in cardiac endothelial cells and macrophages. MVP-associated blood proteins are involved in the renewal-polarization of macrophages and regulation of adaptive immune response. Cytokine activity profiling and digital cell quantification show in MVP a shift toward cytokine signature promoting M2 macrophage polarization. Assessment of druggability identify CSF1R, CX3CR1, CCR6, IL33, MMP8, ENPEP and angiotensin receptors as actionable targets in MVP. CONCLUSIONS Hence, integrative analysis identifies networks of candidate molecules and cells involved in immune control and remodeling of the extracellular matrix, which drive the risk of MVP.
Collapse
Affiliation(s)
| | - Mewen Briend
- Genomic Medicine Laboratory, Quebec Heart and Lung Institute, Laval University, Quebec City, QC, Canada
| | - Mame Sokhna Sylla
- Genomic Medicine Laboratory, Quebec Heart and Lung Institute, Laval University, Quebec City, QC, Canada
| | - Samuel Mathieu
- Genomic Medicine Laboratory, Quebec Heart and Lung Institute, Laval University, Quebec City, QC, Canada
| | - Anne Rufiange
- Genomic Medicine Laboratory, Quebec Heart and Lung Institute, Laval University, Quebec City, QC, Canada
| | - Yohan Bossé
- Department of Molecular Medicine, Laval University, Quebec City, QC, Canada
| | - Patrick Mathieu
- Genomic Medicine Laboratory, Quebec Heart and Lung Institute, Laval University, Quebec City, QC, Canada.
- Department of Surgery, Laval University, Quebec City, QC, Canada.
| |
Collapse
|
3
|
Wang L, Lu Y, Li D, Zhou Y, Yu L, Mesa Eguiagaray I, Campbell H, Li X, Theodoratou E. The landscape of the methodology in drug repurposing using human genomic data: a systematic review. Brief Bioinform 2024; 25:bbad527. [PMID: 38279645 PMCID: PMC10818097 DOI: 10.1093/bib/bbad527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/24/2023] [Accepted: 12/19/2023] [Indexed: 01/28/2024] Open
Abstract
The process of drug development is expensive and time-consuming. In contrast, drug repurposing can be introduced to clinical practice more quickly and at a reduced cost. Over the last decade, there has been a significant expansion of large biobanks that link genomic data to electronic health record data, public availability of various databases containing biological and clinical information and rapid development of novel methodologies and algorithms in integrating different sources of data. This review aims to provide a thorough summary of different strategies that utilize genomic data to seek drug-repositioning opportunities. We searched MEDLINE and EMBASE databases to identify eligible studies up until 1 May 2023, with a total of 102 studies finally included after two-step parallel screening. We summarized commonly used strategies for drug repurposing, including Mendelian randomization, multi-omic-based and network-based studies and illustrated each strategy with examples, as well as the data sources implemented. By leveraging existing knowledge and infrastructure to expedite the drug discovery process and reduce costs, drug repurposing potentially identifies new therapeutic uses for approved drugs in a more efficient and targeted manner. However, technical challenges when integrating different types of data and biased or incomplete understanding of drug interactions are important hindrances that cannot be disregarded in the pursuit of identifying novel therapeutic applications. This review offers an overview of drug repurposing methodologies, providing valuable insights and guiding future directions for advancing drug repurposing studies.
Collapse
Affiliation(s)
- Lijuan Wang
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, UK
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ying Lu
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Doudou Li
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yajing Zhou
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lili Yu
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ines Mesa Eguiagaray
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, UK
| | - Harry Campbell
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, UK
| | - Xue Li
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Evropi Theodoratou
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, UK
- Cancer Research UK Edinburgh Centre, The University of Edinburgh MRC Institute of Genetics and Cancer, Edinburgh, UK
| |
Collapse
|
4
|
Long X, Yuan X, Du J. Single-cell and spatial transcriptomics: Advances in heart development and disease applications. Comput Struct Biotechnol J 2023; 21:2717-2731. [PMID: 37181659 PMCID: PMC10173363 DOI: 10.1016/j.csbj.2023.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/11/2023] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Current transcriptomics technologies, including bulk RNA-seq, single-cell RNA sequencing (scRNA-seq), single-nucleus RNA-sequencing (snRNA-seq), and spatial transcriptomics (ST), provide novel insights into the spatial and temporal dynamics of gene expression during cardiac development and disease processes. Cardiac development is a highly sophisticated process involving the regulation of numerous key genes and signaling pathways at specific anatomical sites and developmental stages. Exploring the cell biological mechanisms involved in cardiogenesis also contributes to congenital heart disease research. Meanwhile, the severity of distinct heart diseases, such as coronary heart disease, valvular disease, cardiomyopathy, and heart failure, is associated with cellular transcriptional heterogeneity and phenotypic alteration. Integrating transcriptomic technologies in the clinical diagnosis and treatment of heart diseases will aid in advancing precision medicine. In this review, we summarize applications of scRNA-seq and ST in the cardiac field, including organogenesis and clinical diseases, and provide insights into the promise of single-cell and spatial transcriptomics in translational research and precision medicine.
Collapse
Affiliation(s)
- Xianglin Long
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xin Yuan
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
5
|
Ke Y, Jian-yuan H, Ping Z, Yue W, Na X, Jian Y, Kai-xuan L, Yi-fan S, Han-bin L, Rong L. The progressive application of single-cell RNA sequencing technology in cardiovascular diseases. Biomed Pharmacother 2022; 154:113604. [DOI: 10.1016/j.biopha.2022.113604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/20/2022] [Accepted: 08/23/2022] [Indexed: 11/02/2022] Open
|
6
|
Chignon A, Mathieu S, Rufiange A, Argaud D, Voisine P, Bossé Y, Arsenault BJ, Thériault S, Mathieu P. Enhancer promoter interactome and Mendelian randomization identify network of druggable vascular genes in coronary artery disease. Hum Genomics 2022; 16:8. [PMID: 35246263 PMCID: PMC8895522 DOI: 10.1186/s40246-022-00381-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 02/17/2022] [Indexed: 11/14/2022] Open
Abstract
Coronary artery disease (CAD) is a multifactorial disorder, which is partly heritable. Herein, we implemented a mapping of CAD-associated candidate genes by using genome-wide enhancer-promoter conformation (H3K27ac-HiChIP) and expression quantitative trait loci (eQTL). Enhancer-promoter anchor loops from human coronary artery smooth muscle cells (HCASMC) explained 22% of the heritability for CAD. 3D enhancer-promoter genome mapping of CAD-genes in HCASMC was enriched in vascular eQTL genes. By using colocalization and Mendelian randomization analyses, we identified 58 causal candidate vascular genes including some druggable targets (MAP3K11, CAMK1D, PDGFD, IPO9 and CETP). A network analysis of causal candidate genes was enriched in TGF beta and MAPK pathways. The pharmacologic inhibition of causal candidate gene MAP3K11 in vascular SMC reduced the expression of athero-relevant genes and lowered cell migration, a cardinal process in CAD. Genes connected to enhancers are enriched in vascular eQTL and druggable genes causally associated with CAD.
Collapse
Affiliation(s)
- Arnaud Chignon
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Institut de Cardiologie Et de Pneumologie de Québec, Quebec Heart and Lung Institute/Research Center, Laval University, 2725 Chemin Ste-Foy, Québec, QC, G1V-4G5, Canada
| | - Samuel Mathieu
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Institut de Cardiologie Et de Pneumologie de Québec, Quebec Heart and Lung Institute/Research Center, Laval University, 2725 Chemin Ste-Foy, Québec, QC, G1V-4G5, Canada
| | - Anne Rufiange
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Institut de Cardiologie Et de Pneumologie de Québec, Quebec Heart and Lung Institute/Research Center, Laval University, 2725 Chemin Ste-Foy, Québec, QC, G1V-4G5, Canada
| | - Déborah Argaud
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Institut de Cardiologie Et de Pneumologie de Québec, Quebec Heart and Lung Institute/Research Center, Laval University, 2725 Chemin Ste-Foy, Québec, QC, G1V-4G5, Canada
| | | | - Yohan Bossé
- Department of Molecular Medicine, Laval University, Quebec, Canada
| | | | - Sébastien Thériault
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Quebec, Canada
| | - Patrick Mathieu
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Institut de Cardiologie Et de Pneumologie de Québec, Quebec Heart and Lung Institute/Research Center, Laval University, 2725 Chemin Ste-Foy, Québec, QC, G1V-4G5, Canada. .,Department of Surgery, Laval University, Quebec, Canada.
| |
Collapse
|
7
|
Tejada-Martinez D, Avelar RA, Lopes I, Zhang B, Novoa G, de Magalhães JP, Trizzino M. Positive Selection and Enhancer Evolution Shaped Lifespan and Body Mass in Great Apes. Mol Biol Evol 2022; 39:msab369. [PMID: 34971383 PMCID: PMC8837823 DOI: 10.1093/molbev/msab369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Within primates, the great apes are outliers both in terms of body size and lifespan, since they include the largest and longest-lived species in the order. Yet, the molecular bases underlying such features are poorly understood. Here, we leveraged an integrated approach to investigate multiple sources of molecular variation across primates, focusing on over 10,000 genes, including approximately 1,500 previously associated with lifespan, and additional approximately 9,000 for which an association with longevity has never been suggested. We analyzed dN/dS rates, positive selection, gene expression (RNA-seq), and gene regulation (ChIP-seq). By analyzing the correlation between dN/dS, maximum lifespan, and body mass, we identified 276 genes whose rate of evolution positively correlates with maximum lifespan in primates. Further, we identified five genes, important for tumor suppression, adaptive immunity, metastasis, and inflammation, under positive selection exclusively in the great ape lineage. RNA-seq data, generated from the liver of six species representing all the primate lineages, revealed that 8% of approximately 1,500 genes previously associated with longevity are differentially expressed in apes relative to other primates. Importantly, by integrating RNA-seq with ChIP-seq for H3K27ac (which marks active enhancers), we show that the differentially expressed longevity genes are significantly more likely than expected to be located near a novel "ape-specific" enhancer. Moreover, these particular ape-specific enhancers are enriched for young transposable elements, and specifically SINE-Vntr-Alus. In summary, we demonstrate that multiple evolutionary forces have contributed to the evolution of lifespan and body size in primates.
Collapse
Affiliation(s)
- Daniela Tejada-Martinez
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Roberto A Avelar
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Inês Lopes
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Bruce Zhang
- Institute of Healthy Ageing, and Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - Guy Novoa
- Department of Structure of Macromolecules, Centro Nacional de Biotecnología—CSIC, Madrid, Spain
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Marco Trizzino
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
8
|
Hollander M, Do T, Will T, Helms V. Detecting Rewiring Events in Protein-Protein Interaction Networks Based on Transcriptomic Data. FRONTIERS IN BIOINFORMATICS 2021; 1:724297. [PMID: 36303788 PMCID: PMC9581068 DOI: 10.3389/fbinf.2021.724297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/23/2021] [Indexed: 12/25/2022] Open
Abstract
Proteins rarely carry out their cellular functions in isolation. Instead, eukaryotic proteins engage in about six interactions with other proteins on average. The aggregated protein interactome of an organism forms a “hairy ball”-type protein-protein interaction (PPI) network. Yet, in a typical human cell, only about half of all proteins are expressed at a particular time. Hence, it has become common practice to prune the full PPI network to the subset of expressed proteins. If RNAseq data is available, one can further resolve the specific protein isoforms present in a cell or tissue. Here, we review various approaches, software tools and webservices that enable users to construct context-specific or tissue-specific PPI networks and how these are rewired between two cellular conditions. We illustrate their different functionalities on the example of the interactions involving the human TNR6 protein. In an outlook, we describe how PPI networks may be integrated with epigenetic data or with data on the activity of splicing factors.
Collapse
|