1
|
Mukherjee K, Khan MSA, Howland JG, Xiao C. Glucagon-like Peptide-2 Acts Partially Through Central GLP-2R and MC4R in Mobilizing Stored Lipids from the Intestine. Nutrients 2025; 17:1416. [PMID: 40362725 PMCID: PMC12073721 DOI: 10.3390/nu17091416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/19/2025] [Accepted: 04/20/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Glucagon-like peptide-2 (GLP-2) is a gut hormone secreted in response to nutrient intake and regulates lipid metabolism in the gut. The present study aims to elucidate the underlying mechanism of GLP-2 in stimulating gut lipid secretion in the fasted state by testing whether GLP-2 signals through the brain's GLP-2 receptor and melanocortin 4 receptor (MC4R). Methods: Sprague-Dawley rats were implanted with a mesenteric lymph duct cannula for measuring gut lipid secretion and an intracerebroventricular cannula for infusion of a GLP-2R antagonist (GLP-2(11-33)), an MC4R antagonist (SHU9119), or saline as a control. The rat received a lipid infusion into the small intestine and a peritoneal injection of GLP-2 five hours later. Results: Brain administration of a GLP-2R antagonist or an MC4R antagonist attenuated the stimulatory effects of peripheral GLP-2 on lymph triglyceride output. These effects were associated with differential changes in the expression of key genes in jejunal endothelial cells, smooth muscle cells, and neuronal cells. Conclusions: These results support the involvement of central GLP-2R and MC4R in a neural pathway for GLP-2 to mobilize lipids stored in the gut during the post-absorptive state.
Collapse
Affiliation(s)
| | | | | | - Changting Xiao
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada; (K.M.); (M.S.A.K.); (J.G.H.)
| |
Collapse
|
2
|
Chadwick J, Hinterberg MA, Asselbergs FW, Biegel H, Boersma E, Cappola TP, Chirinos JA, Coresh J, Ganz P, Gordon DA, Kureshi N, Loupey KM, Orlenko A, Ostroff R, Sampson L, Shrestha S, Sweitzer NK, Williams SA, Zhao L, Kardys I, Lanfear DE. Harnessing the Plasma Proteome to Predict Mortality in Heart Failure Subpopulations. Circ Heart Fail 2025; 18:e011208. [PMID: 40052265 PMCID: PMC11995852 DOI: 10.1161/circheartfailure.123.011208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/16/2025] [Accepted: 01/29/2025] [Indexed: 03/30/2025]
Abstract
BACKGROUND We derived and validated proteomic risk scores (PRSs) for heart failure (HF) prognosis that provide absolute risk estimates for all-cause mortality within 1 year. METHODS Plasma samples from individuals with HF with reduced ejection fraction (HFrEF; ejection fraction <40%; training/validation n=1247/762) and preserved ejection fraction (HFpEF; ejection fraction ≥50%; training/validation n=725/785) from 3 independent studies were run on the SomaScan Assay measuring ≈5000 proteins. Machine learning techniques resulted in unique 17- and 14-protein models for HFrEF and HFpEF that predict 1-year mortality. Discrimination was assessed via C-index and 1-year area under the curve (AUC), and survival curves were visualized. PRSs were also compared with Meta-Analysis Global Group in Chronic HF (MAGGIC) score and NT-proBNP (N-terminal pro-B-type natriuretic peptide) measurements and further assessed for sensitivity to disease progression in longitudinal samples (HFrEF: n=396; 1107 samples; HFpEF: n=175; 350 samples). RESULTS In validation, the HFpEF PRS performed significantly better (P≤0.1) for mortality prediction (C-index, 0.79; AUC, 0.82) than MAGGIC (C-index, 0.71; AUC, 0.74) and NT-proBNP (PRS C-index, 0.76 and AUC, 0.81 versus NT-proBNP C-index, 0.72 and AUC, 0.76). The HFrEF PRS performed comparably to MAGGIC (PRS C-index, 0.76 and AUC, 0.83 versus MAGGIC C-index, 0.75 and AUC, 0.84) but had a significantly better C-Index (P=0.026) than NT-proBNP (PRS C-index, 0.75 and AUC, 0.78 versus NT-proBNP C-index, 0.73 and AUC, 0.77). PRS included known HF pathophysiology biomarkers (93%) and novel proteins (7%). Longitudinal assessment revealed that HFrEF and HFpEF PRSs were higher and increased more over time in individuals who experienced a fatal event during follow-up. CONCLUSIONS PRSs can provide valid, accurate, and dynamic prognostic estimates for patients with HF. This approach has the potential to improve longitudinal monitoring of patients and facilitate personalized care.
Collapse
Affiliation(s)
- Jessica Chadwick
- Departments of Clinical Research and Development (J. Chadwick, R.O., K.M.L., S.A.W.), SomaLogic Operating Co Inc, Boulder, CO
| | - Michael A. Hinterberg
- Bioinformatics (M.A.H., H.B., N.K., L.S., S.S.), SomaLogic Operating Co Inc, Boulder, CO
| | - Folkert W. Asselbergs
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands (F.W.A.)
- Health Data Research UK and Institute of Health Informatics, University College London, United Kingdom (F.W.A.)
| | - Hannah Biegel
- Bioinformatics (M.A.H., H.B., N.K., L.S., S.S.), SomaLogic Operating Co Inc, Boulder, CO
| | - Eric Boersma
- Erasmus MC, University Medical Center Rotterdam, the Netherlands (E.B., I.K.)
| | - Thomas P. Cappola
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia (T.P.C.)
| | - Julio A. Chirinos
- University of Pennsylvania Perelman School of Medicine, Philadelphia (J.A.C.)
| | | | - Peter Ganz
- Division of Cardiology, Zuckerberg San Francisco General Hospital and Department of Medicine, University of California, San Francisco (P.G.)
| | | | - Natasha Kureshi
- Bioinformatics (M.A.H., H.B., N.K., L.S., S.S.), SomaLogic Operating Co Inc, Boulder, CO
| | - Kelsey M. Loupey
- Departments of Clinical Research and Development (J. Chadwick, R.O., K.M.L., S.A.W.), SomaLogic Operating Co Inc, Boulder, CO
| | - Alena Orlenko
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA (A.O.)
| | - Rachel Ostroff
- Departments of Clinical Research and Development (J. Chadwick, R.O., K.M.L., S.A.W.), SomaLogic Operating Co Inc, Boulder, CO
| | - Laura Sampson
- Bioinformatics (M.A.H., H.B., N.K., L.S., S.S.), SomaLogic Operating Co Inc, Boulder, CO
| | - Sama Shrestha
- Bioinformatics (M.A.H., H.B., N.K., L.S., S.S.), SomaLogic Operating Co Inc, Boulder, CO
| | | | - Stephen A. Williams
- Departments of Clinical Research and Development (J. Chadwick, R.O., K.M.L., S.A.W.), SomaLogic Operating Co Inc, Boulder, CO
| | - Lei Zhao
- Bristol Myers Squibb, Princeton, NJ (D.A.G., L.Z.)
| | - Isabella Kardys
- Erasmus MC, University Medical Center Rotterdam, the Netherlands (E.B., I.K.)
| | - David E. Lanfear
- Center for Individualized and Genomic Medicine Research, Henry Ford Hospital, Detroit, MI (D.E.L.)
| |
Collapse
|
3
|
Xie X, Huang M, Ma S, Xin Q, Wang Y, Hu L, Zhao H, Li P, Liu M, Yuan R, Miao Y, Zhu Y, Cong W. The role of long non-coding RNAs in cardiovascular diseases: A comprehensive review. Noncoding RNA Res 2025; 11:158-187. [PMID: 39896344 PMCID: PMC11783329 DOI: 10.1016/j.ncrna.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide, posing significant challenges to healthcare systems. Despite advances in medical interventions, the molecular mechanisms underlying CVDs are not yet fully understood. For decades, protein-coding genes have been the focus of CVD research. However, recent advances in genomics have highlighted the importance of long non-coding RNAs (lncRNAs) in cardiovascular health and disease. Changes in lncRNA expression specific to tissues may result from various internal or external factors, leading to tissue damage, organ dysfunction, and disease. In this review, we provide a comprehensive discussion of the regulatory mechanisms underlying lncRNAs and their roles in the pathogenesis and progression of CVDs, such as coronary heart disease, atherosclerosis, heart failure, arrhythmias, cardiomyopathies, and diabetic cardiomyopathy, to explore their potential as therapeutic targets and diagnostic biomarkers.
Collapse
Affiliation(s)
- Xuena Xie
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Meiwen Huang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Shudong Ma
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, China
| | - Qiqi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yuying Wang
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Lantian Hu
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Pengqi Li
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Mei Liu
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rong Yuan
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yu Miao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yizhun Zhu
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
| | - Weihong Cong
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| |
Collapse
|
4
|
Hao H, Eberand BM, Larance M, Haltiwanger RS. Protein O-Fucosyltransferases: Biological Functions and Molecular Mechanisms in Mammals. Molecules 2025; 30:1470. [PMID: 40286076 PMCID: PMC11990869 DOI: 10.3390/molecules30071470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Domain-specific O-fucosylation is an unusual type of glycosylation, where the fucose is directly attached to the serine or threonine residues in specific protein domains via an O-linkage. O-fucosylated proteins play critical roles in a wide variety of biological events and hold important therapeutic values, with the most studied being the Notch receptors and ADAMTS proteins. O-fucose glycans modulate the function of the proteins they modify and are closely associated with various diseases including cancer. In mammals, alongside the well-documented protein O-fucosyltransferase (POFUT) 1-mediated O-fucosylation of epidermal growth factor-like (EGF) repeats and POFUT2-mediated O-fucosylation of thrombospondin type 1 repeats (TSRs), a new type of O-fucosylation was recently identified on elastin microfibril interface (EMI) domains, mediated by POFUT3 and POFUT4 (formerly FUT10 and FUT11). In this review, we present an overview of our current knowledge of O-fucosylation, integrating the latest findings and with a particular focus on its biological functions and molecular mechanisms.
Collapse
Affiliation(s)
- Huilin Hao
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30605, USA;
| | - Benjamin M. Eberand
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; (B.M.E.); (M.L.)
| | - Mark Larance
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; (B.M.E.); (M.L.)
| | | |
Collapse
|
5
|
Lotfinaghsh A, Imam A, Pompian A, Stitziel NO, Javaheri A. Clinical Insights from Proteomics in Heart Failure. Curr Heart Fail Rep 2025; 22:12. [PMID: 40063168 DOI: 10.1007/s11897-025-00698-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/17/2025] [Indexed: 05/13/2025]
Abstract
PURPOSE OF REVIEW The pathophysiology of heart failure (HF), a complex and heterogenous condition, remains to be fully understood. Troponin and b-type natriuretic peptide are the only biomarkers that are utilized in clinical practice for HF clinical management. Recent advances in proteomics present a powerful tool to identify risk markers and ultimately, potential molecular mechanisms underlying HF pathogenesis. Herein, we explore traditional and novel heart biomarkers, highlighting their potential role in the pathogenesis of HF. RECENT FINDINGS Recent proteomic analyses have identified numerous proteins including Galectin-3, sST2, GDF-15, FGF21, Endotrophin, THSB-2, ADAMSTL, SVEP1, and anthracycline that are associated with clinical outcomes in HF. These biomarkers are not presently utilized in HF management but may be useful in the future for prediction of death or HF hospitalization. While traditional biomarkers remain essential, proteomic strategies have revealed additional targets that require further mechanistic exploration. Future research should focus on validating these biomarkers and translating proteomic insights into clinical practice to enhance HF management.
Collapse
Affiliation(s)
- Aynaz Lotfinaghsh
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Adnan Imam
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexander Pompian
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Nathan O Stitziel
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Ali Javaheri
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- John Cochran VA Hospital, St Louis, MO, USA.
| |
Collapse
|
6
|
Maxwell CB, Bhakta N, Denniff MJ, Sandhu JK, Kessler T, Ng LL, Jones DJ, Webb TR, Morris GE. Deep plasma and tissue proteome profiling of knockout mice reveals pathways associated with Svep1 deficiency. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 11:100283. [PMID: 39895831 PMCID: PMC11782998 DOI: 10.1016/j.jmccpl.2025.100283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/26/2024] [Accepted: 01/09/2025] [Indexed: 02/04/2025]
Abstract
Despite strong causal associations with cardiovascular and metabolic disorders including coronary artery disease, hypertension, and type 2 diabetes, as well as a range of other diseases, the exact function of the protein SVEP1 remains largely unknown. Animal models have been employed to investigate how SVEP1 contributes to disease, with a focus on murine models exploring its role in development, cardiometabolic disease and platelet biology. In this study, we aimed to comprehensively phenotype the proteome of Svep1 +/- mice compared to wild-type (WT) littermates using liquid chromatography-tandem mass spectrometry (LC-MS/MS) bottom-up proteomics in plasma, heart, aorta, lung, and kidney to identify dysregulated pathways and biological functions associated with Svep1 deficiency. Our findings reveal that Svep1 deficiency leads to significant proteomic alterations across the mouse, with the highest number of dysregulated proteins observed in plasma and kidney. Key dysregulated proteins in plasma include upregulation of ADGRV1, CDH1, and MYH6, and downregulation of MTIF2 and AKAP13 which, alongside other proteins dysregulated across tissues, indicate disruption in cell adhesion, extracellular matrix organisation, platelet degranulation, and Rho GTPase pathways. Novel findings include significant enrichment of complement cascades in plasma, suggesting dysregulation of innate immune responses and hemostasis due to Svep1 deficiency. Pathways related to chylomicron assembly and lipid metabolism were also enriched. Additionally, we developed a high-throughput quantitative targeted LC-MS/MS assay to measure endogenous levels of murine SVEP1. SVEP1 was detectable in lung homogenate and showed a significant reduction in SVEP1 levels in Svep1 +/- vs. WT, but was not identified in plasma, heart, aorta, or kidney, likely due to expression levels below the assay's detection limit. Overall, this deep phenotyping study provides insight into the systemic impact of Svep1 deficiency.
Collapse
Affiliation(s)
- Colleen B. Maxwell
- Department of Cardiovascular Sciences and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
- Leicester van Geest multiOMICS Facility, Hodgkin Building, University of Leicester, Leicester LE1 9HN, UK
| | - Nikita Bhakta
- Department of Cardiovascular Sciences and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
- Leicester van Geest multiOMICS Facility, Hodgkin Building, University of Leicester, Leicester LE1 9HN, UK
| | - Matthew J. Denniff
- Department of Cardiovascular Sciences and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Jatinderpal K. Sandhu
- Department of Cardiovascular Sciences and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
- Leicester van Geest multiOMICS Facility, Hodgkin Building, University of Leicester, Leicester LE1 9HN, UK
| | - Thorsten Kessler
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, 80636 Munich, Germany
| | - Leong L. Ng
- Department of Cardiovascular Sciences and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
- Leicester van Geest multiOMICS Facility, Hodgkin Building, University of Leicester, Leicester LE1 9HN, UK
| | - Donald J.L. Jones
- Department of Cardiovascular Sciences and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
- Leicester van Geest multiOMICS Facility, Hodgkin Building, University of Leicester, Leicester LE1 9HN, UK
- Leicester Cancer Research Centre, RKCSB, University of Leicester, Leicester LE2 7LX, UK
| | - Tom R. Webb
- Department of Cardiovascular Sciences and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Gavin E. Morris
- Department of Cardiovascular Sciences and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| |
Collapse
|
7
|
Osawa I, Akita K, Hasegawa K, Fifer MA, Tower-Rader A, Reilly MP, Maurer MS, Stitziel NO, Javaheri A, Shimada YJ. Plasma SVEP1 Levels Predict Cardiovascular Events in Hypertrophic Cardiomyopathy Beyond Conventional Clinical Risk Models Including NT-proBNP. Circ Heart Fail 2025; 18:e012343. [PMID: 39831317 PMCID: PMC11835532 DOI: 10.1161/circheartfailure.124.012343] [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: 08/28/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Hypertrophic cardiomyopathy is the most common genetic cardiomyopathy and causes major adverse cardiovascular events (MACE). SVEP1 (Sushi, von Willebrand factor type A, epidermal growth factor, and pentraxin domain containing 1) is a large extracellular matrix protein that is detectable in the plasma. However, it is unknown whether adding plasma SVEP1 levels to clinical predictors including NT-proBNP (N-terminal pro-B-type natriuretic peptide) improves the prognostication in patients with hypertrophic cardiomyopathy. METHODS We performed a multicenter prospective cohort study of 610 patients with hypertrophic cardiomyopathy. The outcome was MACE defined as heart failure hospitalization or cardiac death. In 4 groups stratified by the median levels of SVEP1 and NT-proBNP, we compared the risk of MACE using the Cox proportional hazards model adjusting for 15 clinical predictors. We also developed a Lasso-regularized Cox proportional hazards model to predict time to first MACE by adding SVEP1 to the 15 clinical predictors with or without NT-proBNP and compared the predictive performance based on C statistics using 10-fold cross-validation. RESULTS Even in the low NT-proBNP groups, the high SVEP1 group had higher risks of MACE compared with the low SVEP1 group (adjusted hazard ratio, 4.52 [95% CI, 1.05-19.4]; P=0.042). In predicting time to first MACE, the addition of SVEP1 improved the C statistics of the clinical plus NT-proBNP model (0.87 [0.83-0.91] versus 0.82 [0.78-0.86]; P=0.01). The clinical plus SVEP1 model also outperformed the clinical plus NT-proBNP model (0.86 [0.82-0.91] versus 0.82 [0.78-0.86]; P=0.04). CONCLUSIONS SVEP1 improved the predictive performance of conventional models, including known clinical parameters with or without NT-proBNP, to predict future MACE in patients with hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Itsuki Osawa
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Keitaro Akita
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Michael A. Fifer
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Albree Tower-Rader
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Muredach P. Reilly
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
- Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY
| | - Mathew S. Maurer
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Nathan O. Stitziel
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO
| | - Ali Javaheri
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| | - Yuichi J. Shimada
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
8
|
Gomez GT, Sathyan S, Chen J, Fornage M, Schlosser P, Peng Z, Cordon J, Palta P, Sullivan KJ, Tin A, Windham BG, Gottesman RF, Barzilai N, Milman S, Verghese J, Coresh J, Walker KA. Plasma proteomic characterization of motoric cognitive risk and mild cognitive impairment. Alzheimers Dement 2025; 21:e14429. [PMID: 39887533 PMCID: PMC11848158 DOI: 10.1002/alz.14429] [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: 07/11/2024] [Revised: 10/23/2024] [Accepted: 10/27/2024] [Indexed: 02/01/2025]
Abstract
INTRODUCTION Motoric cognitive risk (MCR) is a pre-dementia syndrome characterized by mobility and cognitive dysfunction. This study conducted a proteome-wide study of MCR and compared the proteomic signatures of MCR to that of mild cognitive impairment (MCI). METHODS Participants were classified as MCR using a memory questionnaire and 4-meter walk. We measured 4877 plasma proteins collected during late-life and midlife. Multivariable logistic regression related each protein to late-life MCR/MCI. MCR-associated proteins were replicated internally at midlife and in an external cohort. RESULTS Proteome-wide analysis (n = 4076) identified 25 MCR-associated proteins. Eight of these proteins remained associated with late-life MCR when measured during midlife. Two proteins (SVEP1 and TAGLN) were externally replicated. Compared to MCI, MCR had a distinct and much stronger proteomic signature enriched for cardiometabolic and immune pathways. DISCUSSION Our findings highlight the divergent biology underlying two pre-dementia syndromes. Metabolic and immune dysfunction may be a primary driver of MCR. HIGHLIGHTS MCR is defined by concurrent cognitive and gait dysfunction. MCR protein biomarkers have key roles in cardiometabolic and vascular function. MCR biomarkers are also associated with cerebrovascular disease and dementia. MCR and MCI demonstrate overlapping but divergent proteomic signatures.
Collapse
Affiliation(s)
- Gabriela T. Gomez
- Department of Internal MedicineMass General BrighamBostonMassachusettsUSA
| | - Sanish Sathyan
- Department of NeurologyAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Jingsha Chen
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular MedicineMcGovern Medical School and Human Genetics Center, School of Public Health, The University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Pascal Schlosser
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Zhongsheng Peng
- Laboratory of Behavioral NeuroscienceNational Institute on AgingBaltimoreMarylandUSA
| | - Jenifer Cordon
- Laboratory of Behavioral NeuroscienceNational Institute on AgingBaltimoreMarylandUSA
| | - Priya Palta
- Gillings School of Global Public HealthUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Kevin J. Sullivan
- Department of MedicineDivision of GeriatricsUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Adrienne Tin
- MIND Center and Division of NephrologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - B. Gwen Windham
- Department of MedicineDivision of GeriatricsUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Rebecca F. Gottesman
- National Institute of Neurological Disorders and StrokeIntramural Research ProgramBethesdaMarylandUSA
| | - Nir Barzilai
- Department of MedicineDepartment of GeneticsInstitute for Aging Research, Albert Einstein College of MedicineBronxNew YorkUSA
| | - Sofiya Milman
- Department of MedicineDepartment of GeneticsInstitute for Aging Research, Albert Einstein College of MedicineBronxNew YorkUSA
| | - Joe Verghese
- Department of NeurologyRenaissance School of MedicineStony BrookNew YorkUSA
| | - Josef Coresh
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Keenan A. Walker
- Laboratory of Behavioral NeuroscienceNational Institute on AgingBaltimoreMarylandUSA
| |
Collapse
|
9
|
Elenbaas JS, Lee PC, Patel V, Stitziel NO. Decoding the Therapeutic Target SVEP1: Harnessing Molecular Trait GWASs to Unravel Mechanisms of Human Disease. Annu Rev Pharmacol Toxicol 2025; 65:131-148. [PMID: 39847464 DOI: 10.1146/annurev-pharmtox-061724-080905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Although human genetics has substantial potential to illuminate novel disease pathways and facilitate drug development, identifying causal variants and deciphering their mechanisms remain challenging. We believe these challenges can be addressed, in part, by creatively repurposing the results of molecular trait genome-wide association studies (GWASs). In this review, we introduce techniques related to molecular GWASs and unconventionally apply them to understanding SVEP1, a human coronary artery disease risk locus. Our analyses highlight SVEP1's causal link to cardiometabolic disease and glaucoma, as well as the surprising discovery of SVEP1 as the first known physiologic ligand for PEAR1, a critical receptor governing platelet reactivity. We further employ these techniques to dissect the interactions between SVEP1, PEAR1, and the Ang/Tie pathway, with therapeutic implications for a constellation of diseases. This review underscores the potential of molecular GWASs to guide drug discovery and unravel the complexities of human health and disease by demonstrating an integrative approach that grounds mechanistic research in human biology.
Collapse
Affiliation(s)
- Jared S Elenbaas
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA;
- Medical Scientist Training Program, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Paul C Lee
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA;
- Medical Scientist Training Program, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Ved Patel
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA;
| | - Nathan O Stitziel
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA;
- Department of Genetics, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
10
|
Duong T, Austin TR, Brody JA, Shojaie A, Battle A, Bader JS, Hong YS, Ballantyne CM, Coresh J, Gerszten RE, Tracy RP, Psaty BM, Sotoodehnia N, Arking DE. Circulating Blood Plasma Profiling Reveals Proteomic Signature and a Causal Role for SVEP1 in Sudden Cardiac Death. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004494. [PMID: 39234668 PMCID: PMC11479847 DOI: 10.1161/circgen.123.004494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Affiliation(s)
- ThuyVy Duong
- McKusick-Nathans Inst, Dept of Genetic Medicine, Johns Hopkins Univ School of Medicine, Baltimore, MD
| | - Thomas R. Austin
- Cardiovascular Health Rsrch Unit, Dept of Medicine, Univ of Washington, Seattle, WA
- Dept of Epidemiology, Univ of Washington, Seattle, WA
| | - Jennifer A. Brody
- Cardiovascular Health Rsrch Unit, Dept of Medicine, Univ of Washington, Seattle, WA
| | - Ali Shojaie
- Dept of Biostatistics, Univ of Washington, Seattle, WA
| | - Alexis Battle
- Dept of Biomedical Engineering, Johns Hopkins Univ, Baltimore, MD
- Dept of Computer Science, Johns Hopkins Univ, Baltimore, MD
| | - Joel S. Bader
- Dept of Biomedical Engineering, Johns Hopkins Univ, Baltimore, MD
| | - Yun Soo Hong
- McKusick-Nathans Inst, Dept of Genetic Medicine, Johns Hopkins Univ School of Medicine, Baltimore, MD
| | | | - Josef Coresh
- Dept of Epidemiology, Johns Hopkins Univ Bloomberg School of Public Health, Baltimore, MD
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Ctr, Boston, MA
| | - Russell P. Tracy
- Dept of Pathology & Laboratory Medicine & Biochemistry, Larner College of Medicine, Univ of Vermont, Burlington, VT
| | - Bruce M. Psaty
- Cardiovascular Health Rsrch Unit, Dept of Medicine, Univ of Washington, Seattle, WA
- Dept of Epidemiology, Univ of Washington, Seattle, WA
- Dept of Health Systems & Population Health, Univ of Washington, Seattle, WA
| | - Nona Sotoodehnia
- Cardiovascular Health Rsrch Unit, Dept of Medicine, Univ of Washington, Seattle, WA
| | - Dan E Arking
- McKusick-Nathans Inst, Dept of Genetic Medicine, Johns Hopkins Univ School of Medicine, Baltimore, MD
| |
Collapse
|
11
|
Dubin RF, Deo R, Ren Y, Wang J, Pico AR, Mychaleckyj JC, Kozlitina J, Arthur V, Lee H, Shah A, Feldman H, Bansal N, Zelnick L, Rao P, Sukul N, Raj DS, Mehta R, Rosas SE, Bhat Z, Weir MR, He J, Chen J, Kansal M, Kimmel PL, Ramachandran VS, Waikar SS, Segal MR, Ganz P. Incident heart failure in chronic kidney disease: proteomics informs biology and risk stratification. Eur Heart J 2024; 45:2752-2767. [PMID: 38757788 PMCID: PMC11313584 DOI: 10.1093/eurheartj/ehae288] [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: 07/30/2023] [Revised: 04/09/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND AND AIMS Incident heart failure (HF) among individuals with chronic kidney disease (CKD) incurs hospitalizations that burden patients and health care systems. There are few preventative therapies, and the Pooled Cohort equations to Prevent Heart Failure (PCP-HF) perform poorly in the setting of CKD. New drug targets and better risk stratification are urgently needed. METHODS In this analysis of incident HF, SomaScan V4.0 (4638 proteins) was analysed in 2906 participants of the Chronic Renal Insufficiency Cohort (CRIC) with validation in the Atherosclerosis Risk in Communities (ARIC) study. The primary outcome was 14-year incident HF (390 events); secondary outcomes included 4-year HF (183 events), HF with reduced ejection fraction (137 events), and HF with preserved ejection fraction (165 events). Mendelian randomization and Gene Ontology were applied to examine causality and pathways. The performance of novel multi-protein risk models was compared to the PCP-HF risk score. RESULTS Over 200 proteins were associated with incident HF after adjustment for estimated glomerular filtration rate at P < 1 × 10-5. After adjustment for covariates including N-terminal pro-B-type natriuretic peptide, 17 proteins remained associated at P < 1 × 10-5. Mendelian randomization associations were found for six proteins, of which four are druggable targets: FCG2B, IGFBP3, CAH6, and ASGR1. For the primary outcome, the C-statistic (95% confidence interval [CI]) for the 48-protein model in CRIC was 0.790 (0.735, 0.844) vs. 0.703 (0.644, 0.762) for the PCP-HF model (P = .001). C-statistic (95% CI) for the protein model in ARIC was 0.747 (0.707, 0.787). CONCLUSIONS Large-scale proteomics reveal novel circulating protein biomarkers and potential mediators of HF in CKD. Proteomic risk models improve upon the PCP-HF risk score in this population.
Collapse
Affiliation(s)
- Ruth F Dubin
- Division of Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, H5.122E, Dallas, TX 75390, USA
| | - Rajat Deo
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yue Ren
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jianqiao Wang
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alexander R Pico
- Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
| | - Josyf C Mychaleckyj
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Julia Kozlitina
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Victoria Arthur
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hongzhe Lee
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amil Shah
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Harold Feldman
- Patient-Centered Outcomes Research Institute, Washington, DC, USA
| | - Nisha Bansal
- Division of Nephrology, University of Washington Medical Center, Seattle, WA, USA
| | - Leila Zelnick
- Division of Nephrology, University of Washington Medical Center, Seattle, WA, USA
| | - Panduranga Rao
- Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Nidhi Sukul
- Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Dominic S Raj
- Division of Kidney Diseases and Hypertension, George Washington University School of Medicine, Washington, DC, USA
| | - Rupal Mehta
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, USA
| | - Sylvia E Rosas
- Joslin Diabetes Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Zeenat Bhat
- Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Matthew R Weir
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jiang He
- Department of Epidemiology, Tulane University, New Orleans, LA, USA
| | - Jing Chen
- Department of Epidemiology, Tulane University, New Orleans, LA, USA
| | - Mayank Kansal
- Division of Cardiology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Paul L Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Vasan S Ramachandran
- University of Texas School of Public Health San Antonio and the University of Texas Health Sciences Center in San Antonio, Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sushrut S Waikar
- Section of Nephrology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Mark R Segal
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Peter Ganz
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
12
|
Casanova R, Walker KA, Justice JN, Anderson A, Duggan MR, Cordon J, Barnard RT, Lu L, Hsu FC, Sedaghat S, Prizment A, Kritchevsky SB, Wagenknecht LE, Hughes TM. Associations of plasma proteomics and age-related outcomes with brain age in a diverse cohort. GeroScience 2024; 46:3861-3873. [PMID: 38438772 PMCID: PMC11226584 DOI: 10.1007/s11357-024-01112-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/26/2024] [Indexed: 03/06/2024] Open
Abstract
Machine learning models are increasingly being used to estimate "brain age" from neuroimaging data. The gap between chronological age and the estimated brain age gap (BAG) is potentially a measure of accelerated and resilient brain aging. Brain age calculated in this fashion has been shown to be associated with mortality, measures of physical function, health, and disease. Here, we estimate the BAG using a voxel-based elastic net regression approach, and then, we investigate its associations with mortality, cognitive status, and measures of health and disease in participants from Atherosclerosis Risk in Communities (ARIC) study who had a brain MRI at visit 5 of the study. Finally, we used the SOMAscan assay containing 4877 proteins to examine the proteomic associations with the MRI-defined BAG. Among N = 1849 participants (age, 76.4 (SD 5.6)), we found that increased values of BAG were strongly associated with increased mortality and increased severity of the cognitive status. Strong associations with mortality persisted when the analyses were performed in cognitively normal participants. In addition, it was strongly associated with BMI, diabetes, measures of physical function, hypertension, prevalent heart disease, and stroke. Finally, we found 33 proteins associated with BAG after a correction for multiple comparisons. The top proteins with positive associations to brain age were growth/differentiation factor 15 (GDF-15), Sushi, von Willebrand factor type A, EGF, and pentraxin domain-containing protein 1 (SEVP 1), matrilysin (MMP7), ADAMTS-like protein 2 (ADAMTS), and heat shock 70 kDa protein 1B (HSPA1B) while EGF-receptor (EGFR), mast/stem-cell-growth-factor-receptor (KIT), coagulation-factor-VII, and cGMP-dependent-protein-kinase-1 (PRKG1) were negatively associated to brain age. Several of these proteins were previously associated with dementia in ARIC. These results suggest that circulating proteins implicated in biological aging, cellular senescence, angiogenesis, and coagulation are associated with a neuroimaging measure of brain aging.
Collapse
Affiliation(s)
- Ramon Casanova
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC, USA.
| | | | - Jamie N Justice
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Andrea Anderson
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC, USA
| | | | | | - Ryan T Barnard
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC, USA
| | - Lingyi Lu
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC, USA
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC, USA
| | - Sanaz Sedaghat
- School of Public Health, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Anna Prizment
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Stephen B Kritchevsky
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Lynne E Wagenknecht
- Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Timothy M Hughes
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
13
|
Patel‐Murray NL, Zhang L, Claggett BL, Xu D, Serrano‐Fernandez P, Healey M, Wandel S, Chen C, Jacob J, Xu H, Turner GM, Chutkow W, Yates DP, O'Donnell CJ, Prescott MF, Lefkowitz M, Gimpelewicz CR, Beste MT, Zhao F, Gou L, Desai AS, Jhund PS, Packer M, Pfeffer MA, Redfield MM, Rouleau JL, Zannad F, Zile MR, McMurray JJV, Mendelson MM, Solomon SD, Cunningham JW. Aptamer Proteomics for Biomarker Discovery in Heart Failure With Preserved Ejection Fraction: The PARAGON-HF Proteomic Substudy. J Am Heart Assoc 2024; 13:e033544. [PMID: 38904251 PMCID: PMC11255700 DOI: 10.1161/jaha.123.033544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/15/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Prognostic markers and biological pathways linked to detrimental clinical outcomes in heart failure with preserved ejection fraction (HFpEF) remain incompletely defined. METHODS AND RESULTS We measured serum levels of 4123 unique proteins in 1117 patients with HFpEF enrolled in the PARAGON-HF (Efficacy and Safety of LCZ696 Compared to Valsartan, on Morbidity and Mortality in Heart Failure Patients With Preserved Ejection Fraction) trial using a modified aptamer proteomic assay. Baseline circulating protein concentrations significantly associated with the primary end point and the timing and occurrence of total heart failure hospitalization and cardiovascular death were identified by recurrent events regression, accounting for multiple testing, adjusted for age, sex, treatment, and anticoagulant use, and compared with published analyses in 2515 patients with heart failure with reduced ejection fraction from the PARADIGM-HF (Prospective Comparison of ARNI With ACEI to Determine Impact on Global Mortality and Morbidity in Heart Failure) and ATMOSPHERE (Efficacy and Safety of Aliskiren and Aliskiren/Enalapril Combination on Morbidity-Mortality in Patients With Chronic Heart Failure) clinical trials. We identified 288 proteins that were robustly associated with the risk of heart failure hospitalization and cardiovascular death in patients with HFpEF. The baseline proteins most strongly related to outcomes included B2M (β-2 microglobulin), TIMP1 (tissue inhibitor of matrix metalloproteinase 1), SERPINA4 (serpin family A member 4), and SVEP1 (sushi, von Willebrand factor type A, EGF, and pentraxin domain containing 1). Overall, the protein-outcome associations in patients with HFpEF did not markedly differ as compared with patients with heart failure with reduced ejection fraction. A proteomic risk score derived in patients with HFpEF was not superior to a previous proteomic score derived in heart failure with reduced ejection fraction nor to clinical risk factors, NT-proBNP (N-terminal pro-B-type natriuretic peptide), or high-sensitivity cardiac troponin. CONCLUSIONS Numerous serum proteins linked to metabolic, coagulation, and extracellular matrix regulatory pathways were associated with worse HFpEF prognosis in the PARAGON-HF proteomic substudy. Our results demonstrate substantial similarities among serum proteomic risk markers for heart failure hospitalization and cardiovascular death when comparing clinical trial participants with heart failure across the ejection fraction spectrum. REGISTRATION URL: https://www.clinicaltrials.gov; Unique Identifiers: NCT01920711, NCT01035255, NCT00853658.
Collapse
Affiliation(s)
| | - Luqing Zhang
- Novartis Institutes for Biomedical ResearchCambridgeMAUSA
| | - Brian L. Claggett
- Cardiovascular Division, Brigham and Women’s HospitalHarvard Medical SchoolBostonMAUSA
| | - Dongchu Xu
- Cardiovascular Division, Brigham and Women’s HospitalHarvard Medical SchoolBostonMAUSA
| | | | | | | | | | - Jaison Jacob
- Novartis Institutes for Biomedical ResearchCambridgeMAUSA
| | - Huilei Xu
- Novartis Institutes for Biomedical ResearchCambridgeMAUSA
| | | | | | | | | | | | | | | | | | - Faye Zhao
- Novartis Institutes for Biomedical ResearchCambridgeMAUSA
| | - Liangke Gou
- Novartis Pharmaceuticals CorporationEast HanoverNJUSA
| | - Akshay S. Desai
- Cardiovascular Division, Brigham and Women’s HospitalHarvard Medical SchoolBostonMAUSA
| | - Pardeep S. Jhund
- BHF Glasgow Cardiovascular Research Center, School of Cardiovascular and Metabolic HealthUniversity of GlasgowGlasgowUK
| | | | - Marc A. Pfeffer
- Cardiovascular Division, Brigham and Women’s HospitalHarvard Medical SchoolBostonMAUSA
| | | | - Jean L. Rouleau
- Institut de Cardiologie de MontréalUniversité de MontréalQBCanada
| | - Faiez Zannad
- Inserm, Centre d’Investigation Clinique Plurithématique 1433, U1116, CHRU de Nancy, F‐CRIN INI‐CRCTUniversité de LorraineNancyFrance
| | - Michael R. Zile
- Medical University of South Carolina and RHJ Department of Veterans Administration Medical CenterCharlestonSCUSA
| | - John J. V. McMurray
- BHF Glasgow Cardiovascular Research Center, School of Cardiovascular and Metabolic HealthUniversity of GlasgowGlasgowUK
| | | | - Scott D. Solomon
- Cardiovascular Division, Brigham and Women’s HospitalHarvard Medical SchoolBostonMAUSA
| | | |
Collapse
|
14
|
Perry AS, Farber-Eger E, Gonzales T, Tanaka T, Robbins JM, Murthy VL, Stolze LK, Zhao S, Huang S, Colangelo LA, Deng S, Hou L, Lloyd-Jones DM, Walker KA, Ferrucci L, Watts EL, Barber JL, Rao P, Mi MY, Gabriel KP, Hornikel B, Sidney S, Houstis N, Lewis GD, Liu GY, Thyagarajan B, Khan SS, Choi B, Washko G, Kalhan R, Wareham N, Bouchard C, Sarzynski MA, Gerszten RE, Brage S, Wells QS, Nayor M, Shah RV. Proteomic analysis of cardiorespiratory fitness for prediction of mortality and multisystem disease risks. Nat Med 2024; 30:1711-1721. [PMID: 38834850 PMCID: PMC11186767 DOI: 10.1038/s41591-024-03039-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 04/30/2024] [Indexed: 06/06/2024]
Abstract
Despite the wide effects of cardiorespiratory fitness (CRF) on metabolic, cardiovascular, pulmonary and neurological health, challenges in the feasibility and reproducibility of CRF measurements have impeded its use for clinical decision-making. Here we link proteomic profiles to CRF in 14,145 individuals across four international cohorts with diverse CRF ascertainment methods to establish, validate and characterize a proteomic CRF score. In a cohort of around 22,000 individuals in the UK Biobank, a proteomic CRF score was associated with a reduced risk of all-cause mortality (unadjusted hazard ratio 0.50 (95% confidence interval 0.48-0.52) per 1 s.d. increase). The proteomic CRF score was also associated with multisystem disease risk and provided risk reclassification and discrimination beyond clinical risk factors, as well as modulating high polygenic risk of certain diseases. Finally, we observed dynamicity of the proteomic CRF score in individuals who undertook a 20-week exercise training program and an association of the score with the degree of the effect of training on CRF, suggesting potential use of the score for personalization of exercise recommendations. These results indicate that population-based proteomics provides biologically relevant molecular readouts of CRF that are additive to genetic risk, potentially modifiable and clinically translatable.
Collapse
Affiliation(s)
- Andrew S Perry
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Eric Farber-Eger
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Tomas Gonzales
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Toshiko Tanaka
- Longtidudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Jeremy M Robbins
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Lindsey K Stolze
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shilin Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shi Huang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Laura A Colangelo
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Shuliang Deng
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Donald M Lloyd-Jones
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Keenan A Walker
- Multimodal Imaging of Neurodegenerative Disease (MIND) Unit, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Luigi Ferrucci
- Longtidudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Eleanor L Watts
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Jacob L Barber
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Prashant Rao
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michael Y Mi
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kelley Pettee Gabriel
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bjoern Hornikel
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Nicholas Houstis
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
| | - Gregory D Lewis
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
| | - Gabrielle Y Liu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California Davis, Sacramento, CA, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minnesota, MN, USA
| | - Sadiya S Khan
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bina Choi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - George Washko
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Ravi Kalhan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nick Wareham
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Claude Bouchard
- Human Genomic Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Mark A Sarzynski
- Department of Exercise Science, University of South Carolina Columbia, Columbia, SC, USA
| | - Robert E Gerszten
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Soren Brage
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Quinn S Wells
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Matthew Nayor
- Sections of Cardiovascular Medicine and Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Ravi V Shah
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
15
|
Lossos C, Brown J, Sheikhbahaei S, Hubben A, Liu SC, McCrae KR, Chaturvedi S, Naik RP, Francischetti IM. Idiopathic multicentric Castleman disease - TAFRO results in high levels of mTOR activator SVEP1, tissue factor, and endotheliopathy. BLOOD VESSELS, THROMBOSIS & HEMOSTASIS 2024; 1:100006. [PMID: 40191177 PMCID: PMC11970923 DOI: 10.1016/j.bvth.2024.100006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Idiopathic multicentric Castleman disease (iMCD) is an inflammatory disease associated with a cytokine storm, activation of the PI3K/AKT/mTOR pathway, coagulopathy, and increased risk of thrombosis. The mechanisms underlying these pathologic processes remain elusive. We studied novel markers of mTOR activation and thrombosis in 1 patient with typical features of iMCD with TAFRO (thrombocytopenia, anasarca, fevers, reticulin myelofibrosis, and organomegaly) syndrome (iMCD-TAFRO). Plasma levels of SVEP1 (Sushi, von Willebrand factor type A, epidermal growth factor, and pentraxin domain-containing 1 protein), a newly identified mTOR activator associated with cardiovascular diseases and dementia, in addition to cytokines, chemokines and components of the coagulation cascade and complement system were evaluated by enzyme-linked immunosorbent assay (ELISA) and arrays. Compared with healthy controls, a 15-fold increase in SVEP1 was observed. High levels of factor VIIa/antithrombin and microparticles expressing functional tissue factor (TF) were detected. The anticoagulants thrombomodulin and soluble endothelial protein C receptor were elevated, indicating shedding from endothelial cells. Plasminogen activator inhibitor 1 was increased, consistent with hypofibrinolysis, whereas high levels of C3b and C5a are in keeping with complement activation. Furthermore, markers of endothelial cell activation (e.g. von Willebrand factor, angiopoietin-2), cell adhesion molecules, and angiogenesis mediators were upregulated. SVEP1 emerges as a potential mechanism of mTOR activation in iMCD-TAFRO, while multiple pathways influence coagulopathy. Immunothrombosis emerges as a potential therapeutic target for iMCD.
Collapse
Affiliation(s)
- Chen Lossos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jenna Brown
- Division of Adult Hematology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sara Sheikhbahaei
- Division of Radiology and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Anne Hubben
- Department of Hematology/Oncology, Taussig Cancer Institute, and Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Sharon C. Liu
- Department of Hematology/Oncology, Taussig Cancer Institute, and Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Keith R. McCrae
- Department of Hematology/Oncology, Taussig Cancer Institute, and Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Shruti Chaturvedi
- Division of Adult Hematology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Rakhi P. Naik
- Division of Adult Hematology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | |
Collapse
|
16
|
Lotfollahzadeh S, Jose A, Zarnaab Shafiq E, El Sherif N, Smith M, Han J, Seta F, Chitalia V. Two methods of isolation of rat aortic smooth muscle cells with high yield. Biol Methods Protoc 2024; 9:bpae038. [PMID: 39006461 PMCID: PMC11244693 DOI: 10.1093/biomethods/bpae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/30/2024] [Accepted: 05/21/2024] [Indexed: 07/16/2024] Open
Abstract
Vascular smooth muscle cells (VSMCs) are an integral part of blood vessels and are the focus of intensive research in vascular biology, translational research, and cardiovascular diseases. Though immortalized vascular smooth muscle cell lines are available, their use is limited, underscoring the need for primary VSMCs. There are several methods for isolating primary cells from mice. However, the isolation method from rat blood vessels requires optimization, given the differences in the aorta of mice and rats. Here we compare two methods for VSMCs isolation from rats: enzymatic digestion and the "block" method. We observed a significantly higher yield of VSMCs using the enzymatic digestion method. We further confirmed that VSMCs expressed well-established VSMC-specific markers (calponin) with both methods and observed the persistence of this marker up to 9 passages, suggesting a continuation of the secretory phenotype of VSMCs. Overall, this work compares two methods and demonstrates a practical and effective method for isolating VSMCs from rat aorta, providing vascular biologists with a valuable and reliable experimental tool.
Collapse
Affiliation(s)
- Saran Lotfollahzadeh
- Renal Section, Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, United States
| | - Asha Jose
- Renal Section, Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, United States
| | - Esha Zarnaab Shafiq
- Renal Section, Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, United States
| | - Nourhan El Sherif
- Department of Biomedical Engineering, College of Engineering, Boston University, Boston, MA 02118, United States
| | - Michael Smith
- Department of Biomedical Engineering, College of Engineering, Boston University, Boston, MA 02118, United States
| | - Jingyan Han
- Whitaker Cardiovascular Institute, Department of Medicine, Vascular Biology Section, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, United States
| | - Francesca Seta
- Whitaker Cardiovascular Institute, Department of Medicine, Vascular Biology Section, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, United States
| | - Vipul Chitalia
- Renal Section, Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, United States
- Veterans Affairs Boston Healthcare System, Boston, MA 02118, United States
- Center of Cross-Organ Vascular Pathology, Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, United States
| |
Collapse
|
17
|
Dib M, Levin MG, Zhao L, Diab A, Wang Z, Ebert C, Salman O, Azzo JD, Gan S, Zamani P, Cohen JB, Gill D, Burgess S, Zagkos L, van Empel V, Richards AM, Doughty R, Rietzschel ER, Kammerhoff K, Kvikstad E, Maranville J, Schafer P, Seiffert DA, Ramirez‐Valle F, Gordon DA, Chang C, Javaheri A, Mann DL, Cappola TP, Chirinos JA. Proteomic Associations of Adverse Outcomes in Human Heart Failure. J Am Heart Assoc 2024; 13:e031154. [PMID: 38420755 PMCID: PMC10944037 DOI: 10.1161/jaha.123.031154] [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: 10/31/2023] [Accepted: 01/16/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Identifying novel molecular drivers of disease progression in heart failure (HF) is a high-priority goal that may provide new therapeutic targets to improve patient outcomes. The authors investigated the relationship between plasma proteins and adverse outcomes in HF and their putative causal role using Mendelian randomization. METHODS AND RESULTS The authors measured 4776 plasma proteins among 1964 participants with HF with a reduced left ventricular ejection fraction enrolled in PHFS (Penn Heart Failure Study). Assessed were the observational relationship between plasma proteins and (1) all-cause death or (2) death or HF-related hospital admission (DHFA). The authors replicated nominally significant associations in the Washington University HF registry (N=1080). Proteins significantly associated with outcomes were the subject of 2-sample Mendelian randomization and colocalization analyses. After correction for multiple testing, 243 and 126 proteins were found to be significantly associated with death and DHFA, respectively. These included small ubiquitin-like modifier 2 (standardized hazard ratio [sHR], 1.56; P<0.0001), growth differentiation factor-15 (sHR, 1.68; P<0.0001) for death, A disintegrin and metalloproteinase with thrombospondin motifs-like protein (sHR, 1.40; P<0.0001), and pulmonary-associated surfactant protein C (sHR, 1.24; P<0.0001) for DHFA. In pathway analyses, top canonical pathways associated with death and DHFA included fibrotic, inflammatory, and coagulation pathways. Genomic analyses provided evidence of nominally significant associations between levels of 6 genetically predicted proteins with DHFA and 11 genetically predicted proteins with death. CONCLUSIONS This study implicates multiple novel proteins in HF and provides preliminary evidence of associations between genetically predicted plasma levels of 17 candidate proteins and the risk for adverse outcomes in human HF.
Collapse
Affiliation(s)
- Marie‐Joe Dib
- Division of Cardiovascular MedicineHospital of the University of PennsylvaniaPhiladelphiaPAUSA
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| | - Michael G. Levin
- Division of Cardiovascular MedicineHospital of the University of PennsylvaniaPhiladelphiaPAUSA
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| | - Lei Zhao
- Bristol‐Myers Squibb CompanyLawrencevilleNJUSA
| | - Ahmed Diab
- Washington University School of MedicineSt. LouisMOUSA
| | | | | | - Oday Salman
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| | - Joe David Azzo
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| | - Sushrima Gan
- Division of Cardiovascular MedicineHospital of the University of PennsylvaniaPhiladelphiaPAUSA
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| | - Payman Zamani
- Division of Cardiovascular MedicineHospital of the University of PennsylvaniaPhiladelphiaPAUSA
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| | - Jordana B. Cohen
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
- Renal‐Electrolyte and Hypertension Division, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public HealthImperial College LondonLondonUnited Kingdom
| | - Stephen Burgess
- MRC Integrative Epidemiology UnitUniversity of BristolUnited Kingdom
- Department of Public Health and Primary CareUniversity of CambridgeUnited Kingdom
| | - Loukas Zagkos
- Department of Epidemiology and Biostatistics, School of Public HealthImperial College LondonLondonUnited Kingdom
| | - Vanessa van Empel
- Department of CardiologyMaastricht University Medical CenterMaastrichtThe Netherlands
| | - A. Mark Richards
- Department of CardiologyMaastricht University Medical CenterMaastrichtThe Netherlands
- Cardiovascular Research InstituteNational University of SingaporeSingapore
| | - Rob Doughty
- Christchurch Heart InstituteUniversity of OtagoChristchurchNew Zealand
| | | | | | | | | | | | | | | | | | | | - Ali Javaheri
- Washington University School of MedicineSt. LouisMOUSA
- John J. Cochran Veterans HospitalSt. LouisMOUSA
| | | | - Thomas P. Cappola
- Division of Cardiovascular MedicineHospital of the University of PennsylvaniaPhiladelphiaPAUSA
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| | - Julio A. Chirinos
- Division of Cardiovascular MedicineHospital of the University of PennsylvaniaPhiladelphiaPAUSA
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| |
Collapse
|
18
|
Jung IH, Stitziel NO. Integrin α9β1 deficiency does not impact the development of atherosclerosis in mice. Heliyon 2024; 10:e25760. [PMID: 38370227 PMCID: PMC10869861 DOI: 10.1016/j.heliyon.2024.e25760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/20/2024] Open
Abstract
Sushi, von Willebrand factor type A, EGF and pentraxin domain containing 1 (SVEP1) is an extracellular matrix protein that causally promotes cardiovascular disease in humans and mice. However, the receptor mediating the effect of SVEP1 on the development of disease remains unclear. We previously demonstrated that depleting either vascular smooth muscle cell (VSMC)- or myeloid cell-derived integrin α9β1, the first receptor that was identified to interact with SVEP1, did not phenocopy the disease-abrogating effect of depleting SVEP1. Due to its wide expression in tissues and cell types, here we extend this line of investigation to definitively determine if integrin α9β1 impacts the development of atherosclerosis. In a mouse model of atherosclerosis, we found that depleting integrin α9β1 in all cells did not alter plaque size or characteristics of plaque complexity when compared to wild type mice. Further, the significant SVEP1-mediated effects on increase in macrophage content and VSMC proliferation within the atherosclerotic plaque were not altered in animals lacking integrin α9β1. Together, these findings strongly suggest that integrin α9β1 is not responsible for mediating the SVEP1-induced promotion of atherosclerosis and support further studies aimed at characterizing other receptors whose interaction with SVEP1 may represent a therapeutically targetable interaction.
Collapse
Affiliation(s)
- In-Hyuk Jung
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Nathan O. Stitziel
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| |
Collapse
|
19
|
Tian Q, Chen JH, Ding Y, Wang XY, Qiu JY, Cao Q, Zhuang LL, Jin R, Zhou GP. EGR1 transcriptionally regulates SVEP1 to promote proliferation and migration in human coronary artery smooth muscle cells. Mol Biol Rep 2024; 51:365. [PMID: 38409611 DOI: 10.1007/s11033-024-09322-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 02/06/2024] [Indexed: 02/28/2024]
Abstract
A low-frequency variant of sushi, von Willebrand factor type A, EGF, and pentraxin domain-containing protein 1 (SVEP1) is associated with the risk of coronary artery disease, as determined by a genome-wide association study. SVEP1 induces vascular smooth muscle cell proliferation and an inflammatory phenotype to promote atherosclerosis. In the present study, qRT‒PCR demonstrated that the mRNA expression of SVEP1 was significantly increased in atherosclerotic plaques compared to normal tissues. Bioinformatics revealed that EGR1 was a transcription factor for SVEP1. The results of the luciferase reporter assay, siRNA interference or overexpression assay, mutational analysis and ChIP confirmed that EGR1 positively regulated the transcriptional activity of SVEP1 by directly binding to its promoter. EGR1 promoted human coronary artery smooth muscle cell (HCASMC) proliferation and migration via SVEP1 in response to oxidized low-density lipoprotein (ox-LDL) treatment. Moreover, the expression level of EGR1 was increased in atherosclerotic plaques and showed a strong linear correlation with the expression of SVEP1. Our findings indicated that EGR1 binding to the promoter region drive SVEP1 transcription to promote HCASMC proliferation and migration.
Collapse
Affiliation(s)
- Qiang Tian
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jia-He Chen
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Ding
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin-Yu Wang
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jia-Yun Qiu
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qian Cao
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li-Li Zhuang
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Jin
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guo-Ping Zhou
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
20
|
Xu W, Goreczny GJ, Forsythe I, Brennan G, Stowell T, Brock K, Capella B, Turner CE. Hic-5 regulates extracellular matrix-associated gene expression and cytokine secretion in cancer associated fibroblasts. Exp Cell Res 2024; 435:113930. [PMID: 38237846 PMCID: PMC10923124 DOI: 10.1016/j.yexcr.2024.113930] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
The focal adhesion protein, Hic-5 plays a key role in promoting extracellular matrix deposition and remodeling by cancer associated fibroblasts within the tumor stroma to promote breast tumor cell invasion. However, whether stromal matrix gene expression is regulated by Hic-5 is still unknown. Utilizing a constitutive Hic-5 knockout, Mouse Mammary Tumor Virus-Polyoma Middle T-Antigen spontaneous breast tumor mouse model, bulk RNAseq analysis was performed on cancer associated fibroblasts isolated from Hic-5 knockout mammary tumors. Functional network analysis highlighted a key role for Hic-5 in extracellular matrix organization, with both structural matrix genes, as well as matrix remodeling genes being differentially expressed in relation to Hic-5 expression. The subcellular distribution of the MRTF-A transcription factor and expression of a subset of MRTF-A responsive genes was also impacted by Hic-5 expression. Additionally, cytokine array analysis of conditioned media from the Hic-5 and Hic-5 knockout cancer associated fibroblasts revealed that Hic-5 is important for the secretion of several key factors that are associated with matrix remodeling, angiogenesis and immune evasion. Together, these data provide further evidence of a central role for Hic-5 expression in cancer associated fibroblasts in regulating the composition and organization of the tumor stroma microenvironment to promote breast tumor progression.
Collapse
Affiliation(s)
- Weiyi Xu
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Gregory J Goreczny
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA; Jnana Therapeutics, Boston, MA, USA
| | - Ian Forsythe
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA; Zymo Research Corp, Huntington Beach, CA, USA
| | - Grant Brennan
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Theresa Stowell
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Katia Brock
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Benjamin Capella
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Christopher E Turner
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
21
|
Azzo JD, Dib MJ, Zagkos L, Zhao L, Wang Z, Chang CP, Ebert C, Salman O, Gan S, Zamani P, Cohen JB, van Empel V, Richards AM, Javaheri A, Mann DL, Rietzschel E, Schafer P, Seiffert DA, Gill D, Burgess S, Ramirez-Valle F, Gordon DA, Cappola TP, Chirinos JA. Proteomic Associations of NT-proBNP (N-Terminal Pro-B-Type Natriuretic Peptide) in Heart Failure With Preserved Ejection Fraction. Circ Heart Fail 2024; 17:e011146. [PMID: 38299345 PMCID: PMC7615693 DOI: 10.1161/circheartfailure.123.011146] [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: 08/16/2023] [Accepted: 11/20/2023] [Indexed: 02/02/2024]
Abstract
BACKGROUND NT-proBNP (N-terminal pro-B-type natriuretic peptide) levels are variably elevated in heart failure with preserved ejection fraction (HFpEF), even in the presence of increased left ventricular filling pressures. NT-proBNP levels are prognostic in HFpEF and have been used as an inclusion criterion for several recent randomized clinical trials. However, the underlying biologic differences between HFpEF participants with high and low NT-proBNP levels remain to be fully understood. METHODS We measured 4928 proteins using an aptamer-based proteomic assay (SOMAScan) in available plasma samples from 2 cohorts: (1) Participants with HFpEF enrolled in the PHFS (Penn Heart Failure Study; n=253); (2) TOPCAT (Treatment of Preserved Cardiac Function Heart Failure With an Aldosterone Antagonist Trial) participants in the Americas (n=218). We assessed the relationship between SOMAScan-derived plasma NT-proBNP and levels of other proteins available in the SOMAScan assay version 4 using robust linear regression, with correction for multiple comparisons, followed by pathway analysis. RESULTS NT-proBNP levels exhibited prominent proteome-wide associations in PHFS and TOPCAT cohorts. Proteins most strongly associated with NT-proBNP in both cohorts included SVEP1 (sushi, von Willebrand factor type-A, epidermal growth factor, and pentraxin domain containing 1; βTOPCAT=0.539; P<0.0001; βPHFS=0.516; P<0.0001) and ANGPT2 (angiopoietin 2; βTOPCAT=0.571; P<0.0001; βPHFS=0.459; P<0.0001). Canonical pathway analysis demonstrated consistent associations with multiple pathways related to fibrosis and inflammation. These included hepatic fibrosis and inhibition of matrix metalloproteases. Analyses using cut points corresponding to estimated quantitative concentrations of 360 pg/mL (and 480 pg/mL in atrial fibrillation) revealed similar proteomic associations. CONCLUSIONS Circulating NT-proBNP levels exhibit prominent proteomic associations in HFpEF. Our findings suggest that higher NT-proBNP levels in HFpEF are a marker of fibrosis and inflammation. These findings will aid the interpretation of NT-proBNP levels in HFpEF and may guide the selection of participants in future HFpEF clinical trials.
Collapse
Affiliation(s)
- Joe David Azzo
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Marie-Joe Dib
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia PA
| | - Loukas Zagkos
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
| | - Lei Zhao
- Bristol-Myers Squibb Company, Lawrenceville, NJ
| | | | | | | | - Oday Salman
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Sushrima Gan
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia PA
| | - Payman Zamani
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia PA
| | - Jordana B. Cohen
- Bristol-Myers Squibb Company, Lawrenceville, NJ
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA
| | - Vanessa van Empel
- Department of Cardiology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - A. Mark Richards
- Cardiovascular Research Institute, National University of Singapore, Singapore
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Ali Javaheri
- Washington University School of Medicine, St. Louis, MO
- John J. Cochran Veterans Hospital, St. Louis, MO
| | | | - Ernst Rietzschel
- Department of Cardiovascular Diseases, Ghent University Hospital, Ghent, Belgium
| | | | | | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
| | - Stephen Burgess
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | | | - Thomas P. Cappola
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia PA
| | - Julio A. Chirinos
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia PA
| |
Collapse
|
22
|
Axelsson GT, Jonmundsson T, Woo Y, Frick EA, Aspelund T, Loureiro JJ, Orth AP, Jennings LL, Gudmundsson G, Emilsson V, Gudmundsdottir V, Gudnason V. Proteomic associations with forced expiratory volume: a Mendelian randomisation study. Respir Res 2024; 25:44. [PMID: 38238732 PMCID: PMC10797790 DOI: 10.1186/s12931-023-02587-z] [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: 07/21/2023] [Accepted: 10/30/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND A decline in forced expiratory volume (FEV1) is a hallmark of respiratory diseases that are an important cause of morbidity among the elderly. While some data exist on biomarkers that are related to FEV1, we sought to do a systematic analysis of causal relations of biomarkers with FEV1. METHODS Data from the population-based AGES-Reykjavik study were used. Serum proteomic measurements were done using 4782 DNA aptamers (SOMAmers). Data from 1479 participants with spirometric data were used to assess the association of SOMAmer measurements with FEV1 using linear regression. Bi-directional two-sample Mendelian randomisation (MR) analyses were done to assess causal relations of observationally associated SOMAmers with FEV1, using genotype and SOMAmer data from 5368 AGES-Reykjavik participants and genetic associations with FEV1 from a publicly available GWAS (n = 400,102). RESULTS In observational analyses, 530 SOMAmers were associated with FEV1 after multiple testing adjustment (FDR < 0.05). The most significant were Retinoic Acid Receptor Responder 2 (RARRES2), R-Spondin 4 (RSPO4) and Alkaline Phosphatase, Placental Like 2 (ALPPL2). Of the 257 SOMAmers with genetic instruments available, eight were associated with FEV1 in MR analyses. Three were directionally consistent with the observational estimate, Thrombospondin 2 (THBS2), Endoplasmic Reticulum Oxidoreductase 1 Beta (ERO1B) and Apolipoprotein M (APOM). THBS2 was further supported by a colocalization analysis. Analyses in the reverse direction, testing whether changes in SOMAmer levels were caused by changes in FEV1, were performed but no significant associations were found after multiple testing adjustments. CONCLUSIONS In summary, this large scale proteogenomic analyses of FEV1 reveals circulating protein markers of FEV1, as well as several proteins with potential causality to lung function.
Collapse
Affiliation(s)
- Gisli Thor Axelsson
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland
- Department of Internal Medicine, Landspitali University Hospital, 101, Reykjavik, Iceland
| | - Thorarinn Jonmundsson
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland
| | - Youngjae Woo
- Novartis Biomedical Research, Cambridge, MA, 02139, USA
| | | | - Thor Aspelund
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland
| | | | - Anthony P Orth
- Novartis Institutes for Biomedical Research, San Diego, CA, 92121, USA
| | | | - Gunnar Gudmundsson
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland
- Department of Respiratory Medicine and Sleep, Landspitali University Hospital, 108, Reykjavik, Iceland
| | - Valur Emilsson
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland
| | - Valborg Gudmundsdottir
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland.
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland.
| | - Vilmundur Gudnason
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland.
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland.
| |
Collapse
|
23
|
Shah AM, Myhre PL, Arthur V, Dorbala P, Rasheed H, Buckley LF, Claggett B, Liu G, Ma J, Nguyen NQ, Matsushita K, Ndumele C, Tin A, Hveem K, Jonasson C, Dalen H, Boerwinkle E, Hoogeveen RC, Ballantyne C, Coresh J, Omland T, Yu B. Large scale plasma proteomics identifies novel proteins and protein networks associated with heart failure development. Nat Commun 2024; 15:528. [PMID: 38225249 PMCID: PMC10789789 DOI: 10.1038/s41467-023-44680-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 12/21/2023] [Indexed: 01/17/2024] Open
Abstract
Heart failure (HF) causes substantial morbidity and mortality but its pathobiology is incompletely understood. The proteome is a promising intermediate phenotype for discovery of novel mechanisms. We measured 4877 plasma proteins in 13,900 HF-free individuals across three analysis sets with diverse age, geography, and HF ascertainment to identify circulating proteins and protein networks associated with HF development. Parallel analyses in Atherosclerosis Risk in Communities study participants in mid-life and late-life and in Trøndelag Health Study participants identified 37 proteins consistently associated with incident HF independent of traditional risk factors. Mendelian randomization supported causal effects of 10 on HF, HF risk factors, or left ventricular size and function, including matricellular (e.g. SPON1, MFAP4), senescence-associated (FSTL3, IGFBP7), and inflammatory (SVEP1, CCL15, ITIH3) proteins. Protein co-regulation network analyses identified 5 modules associated with HF risk, two of which were influenced by genetic variants that implicated trans hotspots within the VTN and CFH genes.
Collapse
Affiliation(s)
- Amil M Shah
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| | - Peder L Myhre
- Akershus University Hospital and K.G. Jebsen Center for Cardiac Biomarkers, University of Oslo, Oslo, Norway
| | - Victoria Arthur
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Pranav Dorbala
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Humaira Rasheed
- Akershus University Hospital and K.G. Jebsen Center for Cardiac Biomarkers, University of Oslo, Oslo, Norway
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Public Health and Nursing, HUNT Research Center, Norwegian University of Science and Technology, Trondheim, Norway
| | - Leo F Buckley
- Department of Pharmacy, Brigham and Women's Hospital, Boston, MA, USA
| | - Brian Claggett
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Guning Liu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Jianzhong Ma
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Ngoc Quynh Nguyen
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Chiadi Ndumele
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Adrienne Tin
- University of Mississippi Medical Center, Jackson, MS, USA
| | - Kristian Hveem
- Department of Public Health and Nursing, HUNT Research Center, Norwegian University of Science and Technology, Trondheim, Norway
| | - Christian Jonasson
- Department of Public Health and Nursing, HUNT Research Center, Norwegian University of Science and Technology, Trondheim, Norway
| | - Håvard Dalen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Cardiology, St Olavs University Hospital, Trondheim, Norway
- Department of Internal Medicine, Levanger Hospital, Levanger, Norway
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Ron C Hoogeveen
- Division of Cardiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Josef Coresh
- Departments of Medicine and Population Health, NYU Langone Health, New York, NY, USA
| | - Torbjørn Omland
- Akershus University Hospital and K.G. Jebsen Center for Cardiac Biomarkers, University of Oslo, Oslo, Norway
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| |
Collapse
|
24
|
Braz CU, Passamonti MM, Khatib H. Characterization of genomic regions escaping epigenetic reprogramming in sheep. ENVIRONMENTAL EPIGENETICS 2023; 10:dvad010. [PMID: 38496251 PMCID: PMC10944287 DOI: 10.1093/eep/dvad010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 12/04/2023] [Accepted: 12/15/2023] [Indexed: 03/19/2024]
Abstract
The mammalian genome undergoes two global epigenetic reprogramming events during the establishment of primordial germ cells and in the pre-implantation embryo after fertilization. These events involve the erasure and re-establishment of DNA methylation marks. However, imprinted genes and transposable elements (TEs) maintain their DNA methylation signatures to ensure normal embryonic development and genome stability. Despite extensive research in mice and humans, there is limited knowledge regarding environmentally induced epigenetic marks that escape epigenetic reprogramming in other species. Therefore, the objective of this study was to examine the characteristics and locations of genomic regions that evade epigenetic reprogramming in sheep, as well as to explore the biological functions of the genes within these regions. In a previous study, we identified 107 transgenerationally inherited differentially methylated cytosines (DMCs) in the F1 and F2 generations in response to a paternal methionine-supplemented diet. These DMCs were found in TEs, non-repetitive regions, and imprinted and non-imprinted genes. Our findings suggest that genomic regions, rather than TEs and imprinted genes, have the propensity to escape reprogramming and serve as potential candidates for transgenerational epigenetic inheritance. Notably, 34 transgenerational methylated genes influenced by paternal nutrition escaped reprogramming, impacting growth, development, male fertility, cardiac disorders, and neurodevelopment. Intriguingly, among these genes, 21 have been associated with neural development and brain disorders, such as autism, schizophrenia, bipolar disease, and intellectual disability. This suggests a potential genetic overlap between brain and infertility disorders. Overall, our study supports the concept of transgenerational epigenetic inheritance of environmentally induced marks in mammals.
Collapse
Affiliation(s)
- Camila U Braz
- Department of Animal Sciences, University of Illinois Urbana–Champaign, Urbana, IL 61801, USA
| | - Matilde Maria Passamonti
- Department of Animal Science, Food and Nutrition, Universit’a Cattolica del Sacro Cuore, Piacenza, 29122, Italy
| | - Hasan Khatib
- Department of Animal and Dairy Sciences, University of Wisconsin–Madison, Madison, WI 53706, USA
| |
Collapse
|
25
|
Ren Y, Ruan P, Segal M, Dobre M, Schelling JR, Banerjee U, Shafi T, Ganz P, Dubin RF, the CRIC Study Investigators. Evaluation of a large-scale aptamer proteomics platform among patients with kidney failure on dialysis. PLoS One 2023; 18:e0293945. [PMID: 38079395 PMCID: PMC10712847 DOI: 10.1371/journal.pone.0293945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/22/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Patients with kidney failure suffer high mortality, and we currently lack markers for risk stratification for these patients. We carried out a quality control study of a modified aptamer assay (SomaScan v.4.0) that measures ~ 5000 proteins, in preparation for a larger study using this platform in cohorts with kidney failure. METHODS Forty participants from the Cardiac, Endothelial Function and Arterial Stiffness in End-Stage Renal Disease (CERES study) were selected to analyze technical and short-term biological variability, orthogonal correlations and differential protein expression in plasma from patients who died during 2.5 year follow-up. Long-term (one year) variability was studied in 421 participants in the Chronic Renal Insufficiency Cohort. We evaluated 4849 aptamers (4607 unique proteins) using data formats including raw data and data formatted using Adaptive Normalization by Maximum Likelihood (ANML), an algorithm developed for SomaScan data in individuals with normal kidney function. RESULTS In ANML format, median[IQR] intra-assay coefficient of variation (CV) was 2.38%[1.76, 3.40] and inter-assay CV was 7.38%[4.61, 13.12]. Short-term within-subject CV was 5.76% [3.35, 9.72]; long-term CV was 8.71%[5.91, 13.37]. Spearman correlations between aptamer and traditional assays for PTH, NT-proBNP, FGF-23 and CRP were all > 0.7. Fold-change (FC) in protein levels among non-survivors, significant after Bonferroni correction, included SVEP1 (FC[95% CI] 2.14 [1.62, 2.82]), keratocan (1.74 [1.40, 2.15]) and LanC-like protein 1 (0.56 [0.45, 0.70]). Compared to raw aptamer data, technical and short-term biological variability in paired samples was lower in ANML-formatted data. ANML formatting had minimal impact on orthogonal correlations with traditional assays or the associations of proteins with the phenotype of mortality. CONCLUSIONS SomaScan had excellent technical variability and low within-subject short-term variability. ANML formatting could facilitate comparison of biomarker results with other studies that utilize this format. We expect SomaScan to provide novel and reproducible information in patients with kidney failure on dialysis.
Collapse
Affiliation(s)
- Yue Ren
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Peifeng Ruan
- Peter O’Donnell Jr School of Public Health, UT Southwestern, Dallas, Texas, United States of America
| | - Mark Segal
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, United States of America
| | - Mirela Dobre
- Division of Nephrology and Hypertension, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
| | - Jeffrey R. Schelling
- Department of Physiology & Biophysics, Case Western Reserve University of School of Medicine, Cleveland, Ohio, United States of America
| | - Upasana Banerjee
- Department of Internal Medicine, Hurley Medical Center/Michigan State University, Flint, Michigan, United States of America
| | - Tariq Shafi
- Division of Kidney Diseases, Hypertension and Transplantation, Houston Methodist Hospital, Houston, Texas, United States of America
| | - Peter Ganz
- Division of Cardiology, University of California, San Francisco, San Francisco, California, United States of America
| | - Ruth F. Dubin
- Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | | |
Collapse
|
26
|
Verdier H, Thomas P, Batista J, Kempster C, McKinney H, Gleadall N, Danesh J, Mumford A, Heemskerk JWM, Ouwehand WH, Downes K, Astle WJ, Turro E. A signature of platelet reactivity in CBC scattergrams reveals genetic predictors of thrombotic disease risk. Blood 2023; 142:1895-1908. [PMID: 37647652 PMCID: PMC10733829 DOI: 10.1182/blood.2023021100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/27/2023] [Accepted: 08/18/2023] [Indexed: 09/01/2023] Open
Abstract
Genetic studies of platelet reactivity (PR) phenotypes may identify novel antiplatelet drug targets. However, such studies have been limited by small sample sizes (n < 5000) because of the complexity of measuring PR. We trained a model to predict PR from complete blood count (CBC) scattergrams. A genome-wide association study of this phenotype in 29 806 blood donors identified 21 distinct associations implicating 20 genes, of which 6 have been identified previously. The effect size estimates were significantly correlated with estimates from a study of flow cytometry-measured PR and a study of a phenotype of in vitro thrombus formation. A genetic score of PR built from the 21 variants was associated with the incidence rates of myocardial infarction and pulmonary embolism. Mendelian randomization analyses showed that PR was causally associated with the risks of coronary artery disease, stroke, and venous thromboembolism. Our approach provides a blueprint for using phenotype imputation to study the determinants of hard-to-measure but biologically important hematological traits.
Collapse
Affiliation(s)
- Hippolyte Verdier
- Institut Pasteur, CNRS UMR 3751, Decision and Bayesian Computation, Université Paris Cité, Paris, France
| | - Patrick Thomas
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Joana Batista
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Carly Kempster
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Harriet McKinney
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Nicholas Gleadall
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Andrew Mumford
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
- South West National Health Service Genomic Medicine Service Alliance, Bristol, United Kingdom
| | | | - Willem H. Ouwehand
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Kate Downes
- Cambridge Genomics Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - William J. Astle
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Medical Research Council Biostatistics Unit, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, United Kingdom
| | - Ernest Turro
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
27
|
Gao G, Zhang H, Ni J, Zhao X, Zhang K, Wang J, Kong X, Wang Q. Insights into genetic diversity and phenotypic variations in domestic geese through comprehensive population and pan-genome analysis. J Anim Sci Biotechnol 2023; 14:150. [PMID: 38001525 PMCID: PMC10675864 DOI: 10.1186/s40104-023-00944-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/06/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Domestic goose breeds are descended from either the Swan goose (Anser cygnoides) or the Greylag goose (Anser anser), exhibiting variations in body size, reproductive performance, egg production, feather color, and other phenotypic traits. Constructing a pan-genome facilitates a thorough identification of genetic variations, thereby deepening our comprehension of the molecular mechanisms underlying genetic diversity and phenotypic variability. RESULTS To comprehensively facilitate population genomic and pan-genomic analyses in geese, we embarked on the task of 659 geese whole genome resequencing data and compiling a database of 155 RNA-seq samples. By constructing the pan-genome for geese, we generated non-reference contigs totaling 612 Mb, unveiling a collection of 2,813 novel genes and pinpointing 15,567 core genes, 1,324 softcore genes, 2,734 shell genes, and 878 cloud genes in goose genomes. Furthermore, we detected an 81.97 Mb genomic region showing signs of genome selection, encompassing the TGFBR2 gene correlated with variations in body weight among geese. Genome-wide association studies utilizing single nucleotide polymorphisms (SNPs) and presence-absence variation revealed significant genomic associations with various goose meat quality, reproductive, and body composition traits. For instance, a gene encoding the SVEP1 protein was linked to carcass oblique length, and a distinct gene-CDS haplotype of the SVEP1 gene exhibited an association with carcass oblique length. Notably, the pan-genome analysis revealed enrichment of variable genes in the "hair follicle maturation" Gene Ontology term, potentially linked to the selection of feather-related traits in geese. A gene presence-absence variation analysis suggested a reduced frequency of genes associated with "regulation of heart contraction" in domesticated geese compared to their wild counterparts. Our study provided novel insights into gene expression features and functions by integrating gene expression patterns across multiple organs and tissues in geese and analyzing population variation. CONCLUSION This accomplishment originates from the discernment of a multitude of selection signals and candidate genes associated with a wide array of traits, thereby markedly enhancing our understanding of the processes underlying domestication and breeding in geese. Moreover, assembling the pan-genome for geese has yielded a comprehensive apprehension of the goose genome, establishing it as an indispensable asset poised to offer innovative viewpoints and make substantial contributions to future geese breeding initiatives.
Collapse
Affiliation(s)
- Guangliang Gao
- Chongqing Academy of Animal Science, Rongchang District, Chongqing, 402460, China
- Livestock and Poultry Multi-Omics Key Laboratory of Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
- Chongqing Engineering Research Center of Goose Genetic Improvement, Rongchang District, Chongqing, 402460, China
| | - Hongmei Zhang
- Department of Cardiovascular Ultrasound and Non-Invasive Cardiology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital,University of Electronic Science and Technology of China, Chengdu, 611731, China
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Jiangping Ni
- JiguangGene Biotechnology Co., Ltd., Nanjing, 210032, China
| | - Xianzhi Zhao
- Chongqing Academy of Animal Science, Rongchang District, Chongqing, 402460, China
- Chongqing Engineering Research Center of Goose Genetic Improvement, Rongchang District, Chongqing, 402460, China
| | - Keshan Zhang
- Chongqing Academy of Animal Science, Rongchang District, Chongqing, 402460, China
- Chongqing Engineering Research Center of Goose Genetic Improvement, Rongchang District, Chongqing, 402460, China
| | - Jian Wang
- Jiangsu Agri-Animal Vocational College, Taizhou, 225300, China
| | - Xiangdong Kong
- JiguangGene Biotechnology Co., Ltd., Nanjing, 210032, China.
| | - Qigui Wang
- Chongqing Academy of Animal Science, Rongchang District, Chongqing, 402460, China.
- Chongqing Engineering Research Center of Goose Genetic Improvement, Rongchang District, Chongqing, 402460, China.
- Present Address: Poultry Science Institute, Chongqing Academy of Animal Science, No. 51 Changzhou Avenue, Rongchang District, Chongqing, 402460, P. R. China.
| |
Collapse
|
28
|
Elenbaas JS, Jung IH, Coler-Reilly A, Lee PC, Alisio A, Stitziel NO. The emerging Janus face of SVEP1 in development and disease. Trends Mol Med 2023; 29:939-950. [PMID: 37673700 PMCID: PMC10592172 DOI: 10.1016/j.molmed.2023.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 09/08/2023]
Abstract
Sushi, von Willebrand factor type A, EGF, and pentraxin domain containing 1 (SVEP1) is a large extracellular matrix protein that is also detected in circulation. Recent plasma proteomic and genomic studies have revealed a large number of associations between SVEP1 and human traits, particularly chronic disease. These include associations with cardiac death and disease, diabetes, platelet traits, glaucoma, dementia, and aging; many of these are causal. Animal models demonstrate that SVEP1 is critical in vascular development and disease, but its molecular and cellular mechanisms remain poorly defined. Future studies should aim to characterize these mechanisms and determine the diagnostic, prognostic, and therapeutic value of measuring or intervening on this enigmatic protein.
Collapse
Affiliation(s)
- Jared S Elenbaas
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Medical Scientist Training Program, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - In-Hyuk Jung
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Ariella Coler-Reilly
- Medical Scientist Training Program, Washington University School of Medicine, Saint Louis, MO 63110, USA; Division of Bone and Mineral Diseases, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Paul C Lee
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Medical Scientist Training Program, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Arturo Alisio
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Nathan O Stitziel
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO 63108, USA; Department of Genetics, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
29
|
He Z, Luo J, Lv M, Li Q, Ke W, Niu X, Zhang Z. Characteristics and evaluation of atherosclerotic plaques: an overview of state-of-the-art techniques. Front Neurol 2023; 14:1159288. [PMID: 37900593 PMCID: PMC10603250 DOI: 10.3389/fneur.2023.1159288] [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: 02/05/2023] [Accepted: 09/28/2023] [Indexed: 10/31/2023] Open
Abstract
Atherosclerosis is an important cause of cerebrovascular and cardiovascular disease (CVD). Lipid infiltration, inflammation, and altered vascular stress are the critical mechanisms that cause atherosclerotic plaque formation. The hallmarks of the progression of atherosclerosis include plaque ulceration, rupture, neovascularization, and intraplaque hemorrhage, all of which are closely associated with the occurrence of CVD. Assessing the severity of atherosclerosis and plaque vulnerability is crucial for the prevention and treatment of CVD. Integrating imaging techniques for evaluating the characteristics of atherosclerotic plaques with computer simulations yields insights into plaque inflammation levels, spatial morphology, and intravascular stress distribution, resulting in a more realistic and accurate estimation of plaque state. Here, we review the characteristics and advancing techniques used to analyze intracranial and extracranial atherosclerotic plaques to provide a comprehensive understanding of atheroma.
Collapse
Affiliation(s)
- Zhiwei He
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiaying Luo
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mengna Lv
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qingwen Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Ke
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xuan Niu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
30
|
Andrews SL, Ghaderi-Najafabadi M, Gong P, Shamkhi N, Carleton L, Schofield C, Kessler T, Samani NJ, Webb TR, Morris GE. SVEP1 influences monocyte to macrophage differentiation via integrin α4β1/α9β1 and Rho/Rac signalling. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119479. [PMID: 37100352 DOI: 10.1016/j.bbamcr.2023.119479] [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: 09/28/2022] [Revised: 03/09/2023] [Accepted: 04/15/2023] [Indexed: 04/28/2023]
Abstract
BACKGROUND The large extracellular matrix protein SVEP1 mediates cell adhesion via integrin α9β1. Recent studies have identified an association between a missense variant in SVEP1 and increased risk of coronary artery disease (CAD) in humans and in mice Svep1 deficiency alters the development of atherosclerotic plaques. However how SVEP1 functionally contributes to CAD pathogenesis is not fully understood. Monocyte recruitment and differentiation to macrophages is a key step in the development of atherosclerosis. Here, we investigated the requirement for SVEP1 in this process. METHODS SVEP1 expression was measured during monocyte-macrophage differentiation in primary monocytes and THP-1 human monocytic cells. SVEP1 knockout THP-1 cell lines and the dual integrin α4β1/α9β1 inhibitor, BOP, were utilised to investigate the effect of these proteins in THP-1 cell adhesion, migration and cell spreading assays. Subsequent activation of downstream integrin signalling intermediaries was quantified by western blotting. RESULTS SVEP1 gene expression increases in monocyte to macrophage differentiation in human primary monocytes and THP-1 cells. Using two SVEP1 knockout THP-1 cells we observed reduction in monocyte adhesion, migration, and cell spreading compared to control cells. Similar results were found with integrin α4β1/α9β1 inhibition. We demonstrate reduced activity of Rho and Rac1 in SVEP1 knockout THP-1 cells. CONCLUSIONS SVEP1 regulates monocyte recruitment and differentiation phenotypes through an integrin α4β1/α9β1 dependent mechanism. GENERAL SIGNIFICANCE These results describe a novel role for SVEP1 in monocyte behaviour relevant to CAD pathophysiology.
Collapse
Affiliation(s)
- S L Andrews
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom.
| | - M Ghaderi-Najafabadi
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom.
| | - P Gong
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom.
| | - N Shamkhi
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom.
| | - L Carleton
- Horizon Discovery Ltd., 8100 Cambridge Research Park, Cambridge CB25 9TL, United Kingdom.
| | - C Schofield
- Horizon Discovery Ltd., 8100 Cambridge Research Park, Cambridge CB25 9TL, United Kingdom.
| | - T Kessler
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany.; German Centre of Cardiovascular Research (DZHK e. V.), Partner Site Munich Heart Alliance, Munich, Germany.
| | - N J Samani
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom.
| | - T R Webb
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom.
| | - G E Morris
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom.
| |
Collapse
|
31
|
Axelsson GT, Jonmundsson T, Woo YJ, Frick EA, Aspelund T, Loureiro JJ, Orth AP, Jennings LL, Gudmundsson G, Emilsson V, Gudmundsdottir V, Gudnason V. Proteomic associations with forced expiratory volume - a Mendelian randomisation study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.30.23292035. [PMID: 37425696 PMCID: PMC10327250 DOI: 10.1101/2023.06.30.23292035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
A decline in forced expiratory volume (FEV1) is a hallmark of obstructive respiratory diseases, an important cause of morbidity among the elderly. While some data exist on biomarkers that are related to FEV1, we sought to do a systematic analysis of causal relations of biomarkers with FEV1. Data from the general population-based AGES-Reykjavik study were used. Proteomic measurements were done using 4,782 DNA aptamers (SOMAmers). Data from 1,648 participants with spirometric data were used to assess the association of SOMAmer measurements with FEV1 using linear regression. Bi-directional Mendelian randomisation (MR) analyses were done to assess causal relations of observationally associated SOMAmers with FEV1, using genotype and SOMAmer data from 5,368 AGES-Reykjavik participants and genetic associations with FEV1 from a publicly available GWAS (n = 400,102). In observational analyses, 473 SOMAmers were associated with FEV1 after multiple testing adjustment. The most significant were R-Spondin 4, Alkaline Phosphatase, Placental Like 2 and Retinoic Acid Receptor Responder 2. Of the 235 SOMAmers with genetic data, eight were associated with FEV1 in MR analyses. Three were directionally consistent with the observational estimate, Thrombospondin 2 (THBS2), Endoplasmic Reticulum Oxidoreductase 1 Beta and Apolipoprotein M. THBS2 was further supported by a colocalization analysis. Analyses in the reverse direction, testing whether changes in SOMAmer levels were caused by changes in FEV1, were performed but no significant associations were found after multiple testing adjustments. In summary, this large scale proteogenomic analyses of FEV1 reveals protein markers of FEV1, as well as several proteins with potential causality to lung function.
Collapse
|
32
|
Deo R, Dubin RF, Ren Y, Murthy AC, Wang J, Zheng H, Zheng Z, Feldman H, Shou H, Coresh J, Grams M, Surapaneni AL, Bhat Z, Cohen JB, Rahman M, He J, Saraf SL, Go AS, Kimmel PL, Vasan RS, Segal MR, Li H, Ganz P. Proteomic cardiovascular risk assessment in chronic kidney disease. Eur Heart J 2023; 44:2095-2110. [PMID: 37014015 PMCID: PMC10281556 DOI: 10.1093/eurheartj/ehad115] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 01/21/2023] [Accepted: 02/16/2023] [Indexed: 04/05/2023] Open
Abstract
AIMS Chronic kidney disease (CKD) is widely prevalent and independently increases cardiovascular risk. Cardiovascular risk prediction tools derived in the general population perform poorly in CKD. Through large-scale proteomics discovery, this study aimed to create more accurate cardiovascular risk models. METHODS AND RESULTS Elastic net regression was used to derive a proteomic risk model for incident cardiovascular risk in 2182 participants from the Chronic Renal Insufficiency Cohort. The model was then validated in 485 participants from the Atherosclerosis Risk in Communities cohort. All participants had CKD and no history of cardiovascular disease at study baseline when ∼5000 proteins were measured. The proteomic risk model, which consisted of 32 proteins, was superior to both the 2013 ACC/AHA Pooled Cohort Equation and a modified Pooled Cohort Equation that included estimated glomerular filtrate rate. The Chronic Renal Insufficiency Cohort internal validation set demonstrated annualized receiver operating characteristic area under the curve values from 1 to 10 years ranging between 0.84 and 0.89 for the protein and 0.70 and 0.73 for the clinical models. Similar findings were observed in the Atherosclerosis Risk in Communities validation cohort. For nearly half of the individual proteins independently associated with cardiovascular risk, Mendelian randomization suggested a causal link to cardiovascular events or risk factors. Pathway analyses revealed enrichment of proteins involved in immunologic function, vascular and neuronal development, and hepatic fibrosis. CONCLUSION In two sizeable populations with CKD, a proteomic risk model for incident cardiovascular disease surpassed clinical risk models recommended in clinical practice, even after including estimated glomerular filtration rate. New biological insights may prioritize the development of therapeutic strategies for cardiovascular risk reduction in the CKD population.
Collapse
Affiliation(s)
- Rajat Deo
- Division of Cardiovascular Medicine, Electrophysiology Section, Perelman School of Medicine at the University of Pennsylvania, One Convention Avenue, Level 2 / City Side, Philadelphia, PA 19104, USA
| | - Ruth F Dubin
- Division of Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Yue Ren
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Ashwin C Murthy
- Division of Cardiovascular Medicine, Electrophysiology Section, Perelman School of Medicine at the University of Pennsylvania, One Convention Avenue, Level 2 / City Side, Philadelphia, PA 19104, USA
| | - Jianqiao Wang
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Haotian Zheng
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Zihe Zheng
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Harold Feldman
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Haochang Shou
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Josef Coresh
- Department of Epidemiology; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins University, 2024 E. Monument Street, Room 2-635, Suite 2-600, Baltimore, MD 21287, USA
| | - Morgan Grams
- Department of Epidemiology; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins University, 2024 E. Monument Street, Room 2-635, Suite 2-600, Baltimore, MD 21287, USA
| | - Aditya L Surapaneni
- Department of Epidemiology; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA
| | - Zeenat Bhat
- Division of Nephrology, University of Michigan, 5100 Brehm Tower, 1000 Wall Street, Ann Arbor, MI 48105, USA
| | - Jordana B Cohen
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
- Renal, Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, 831 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Mahboob Rahman
- Department of Medicine, Case Western Reserve University School of Medicine, 11100 Euclid Avenue, Wearn Bldg. 3 Floor. Rm 352, Cleveland, OH 44106, USA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, SL 18, New Orleans, LA 70112, USA
| | - Santosh L Saraf
- Division of Hematology and Oncology, University of Illinois at Chicago, 1740 West Taylor Street, Chicago, IL 60612, USA
| | - Alan S Go
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA
- Departments of Epidemiology, Biostatistics and Medicine, University of California at San Francisco, San Francisco, CA, USA
| | - Paul L Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Ramachandran S Vasan
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Section of Cardiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Mark R Segal
- Department of Epidemiology and Biostatistics, University of California, 550 16th Street, 2nd Floor, Box #0560, San Francisco, CA 94143, USA
| | - Hongzhe Li
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Peter Ganz
- Division of Cardiology, Zuckerberg San Francisco General Hospital and Department of Medicine, University of California, San Francisco, 1001 Potrero Avenue, 5G1, San Francisco, CA 94110, USA
| |
Collapse
|
33
|
Elenbaas JS, Pudupakkam U, Ashworth KJ, Kang CJ, Patel V, Santana K, Jung IH, Lee PC, Burks KH, Amrute JM, Mecham RP, Halabi CM, Alisio A, Di Paola J, Stitziel NO. SVEP1 is an endogenous ligand for the orphan receptor PEAR1. Nat Commun 2023; 14:850. [PMID: 36792666 PMCID: PMC9932102 DOI: 10.1038/s41467-023-36486-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 01/30/2023] [Indexed: 02/17/2023] Open
Abstract
Sushi, von Willebrand factor type A, EGF and pentraxin domain containing 1 (SVEP1) is an extracellular matrix protein that causally promotes vascular disease and associates with platelet reactivity in humans. Here, using a human genomic and proteomic approach, we identify a high affinity, disease-relevant, and potentially targetable interaction between SVEP1 and the orphan receptor Platelet and Endothelial Aggregation Receptor 1 (PEAR1). This interaction promotes PEAR1 phosphorylation and disease associated AKT/mTOR signaling in vascular cells and platelets. Mice lacking SVEP1 have reduced platelet activation, and exogenous SVEP1 induces PEAR1-dependent activation of platelets. SVEP1 and PEAR1 causally and concordantly relate to platelet phenotypes and cardiovascular disease in humans, as determined by Mendelian Randomization. Targeting this receptor-ligand interaction may be a viable therapeutic strategy to treat or prevent cardiovascular and thrombotic disease.
Collapse
Affiliation(s)
- Jared S Elenbaas
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
- Medical Scientist Training Program, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
| | - Upasana Pudupakkam
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Katrina J Ashworth
- Division of Pediatric Hematology Oncology, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Chul Joo Kang
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, 63108, USA
| | - Ved Patel
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Katherine Santana
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - In-Hyuk Jung
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Paul C Lee
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Medical Scientist Training Program, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Kendall H Burks
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Medical Scientist Training Program, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Junedh M Amrute
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Medical Scientist Training Program, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Robert P Mecham
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Carmen M Halabi
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Department of Pediatrics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Arturo Alisio
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Jorge Di Paola
- Division of Pediatric Hematology Oncology, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Nathan O Stitziel
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, 63108, USA.
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
| |
Collapse
|
34
|
Tarca AL, Romero R, Bhatti G, Gotsch F, Done B, Gudicha DW, Gallo DM, Jung E, Pique-Regi R, Berry SM, Chaiworapongsa T, Gomez-Lopez N. Human Plasma Proteome During Normal Pregnancy. J Proteome Res 2022; 21:2687-2702. [PMID: 36154181 PMCID: PMC10445406 DOI: 10.1021/acs.jproteome.2c00391] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The human plasma proteome is underexplored despite its potential value for monitoring health and disease. Herein, using a recently developed aptamer-based platform, we profiled 7288 proteins in 528 plasma samples from 91 normal pregnancies (Gene Expression Omnibus identifier GSE206454). The coefficient of variation was <20% for 93% of analytes (median 7%), and a cross-platform correlation for selected key angiogenic and anti-angiogenic proteins was significant. Gestational age was associated with changes in 953 proteins, including highly modulated placenta- and decidua-specific proteins, and they were enriched in biological processes including regulation of growth, angiogenesis, immunity, and inflammation. The abundance of proteins corresponding to RNAs specific to populations of cells previously described by single-cell RNA-Seq analysis of the placenta was highly modulated throughout gestation. Furthermore, machine learning-based prediction of gestational age and of time from sampling to term delivery compared favorably with transcriptomic models (mean absolute error of 2 weeks). These results suggested that the plasma proteome may provide a non-invasive readout of placental cellular dynamics and serve as a blueprint for investigating obstetrical disease.
Collapse
Affiliation(s)
- Adi L Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan48202, United States
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan48103, United States
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan48824, United States
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan48202, United States
- Detroit Medical Center, Detroit, Michigan48201, United States
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
| | - Francesca Gotsch
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
| | - Bogdan Done
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
| | - Dereje W Gudicha
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
| | - Dahiana M Gallo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, University of Valle 13, Cali, Valle del Cauca100-00, Colombia
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
| | - Roger Pique-Regi
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan48202, United States
| | - Stanley M Berry
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and, Detroit, Michigan48201, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan48201, United States
| |
Collapse
|
35
|
Morris GE, Denniff MJ, Karamanavi E, Andrews SA, Kostogrys RB, Bountziouka V, Ghaderi‐Najafabadi M, Shamkhi N, McConnell G, Kaiser MA, Carleton L, Schofield C, Kessler T, Rainbow RD, Samani NJ, Webb TR. The integrin ligand SVEP1 regulates GPCR-mediated vasoconstriction via integrins α9β1 and α4β1. Br J Pharmacol 2022; 179:4958-4973. [PMID: 35802072 PMCID: PMC9805129 DOI: 10.1111/bph.15921] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 06/10/2022] [Accepted: 06/27/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE Vascular tone is regulated by the relative contractile state of vascular smooth muscle cells (VSMCs). Several integrins directly modulate VSMC contraction by regulating calcium influx through L-type voltage-gated Ca2+ channels (VGCCs). Genetic variants in ITGA9, which encodes the α9 subunit of integrin α9β1, and SVEP1, a ligand for integrin α9β1, associate with elevated blood pressure; however, neither SVEP1 nor integrin α9β1 has reported roles in vasoregulation. We determined whether SVEP1 and integrin α9β1 can regulate VSMC contraction. EXPERIMENTAL APPROACH SVEP1 and integrin binding were confirmed by immunoprecipitation and cell binding assays. Human induced pluripotent stem cell-derived VSMCs were used in in vitro [Ca2+ ]i studies, and aortas from a Svep1+/- knockout mouse model were used in wire myography to measure vessel contraction. KEY RESULTS We confirmed the ligation of SVEP1 to integrin α9β1 and additionally found SVEP1 to directly bind to integrin α4β1. Inhibition of SVEP1, integrin α4β1 or α9β1 significantly enhanced [Ca2+ ]i levels in isolated VSMCs to Gαq/11 -vasoconstrictors. This response was confirmed in whole vessels where a greater contraction to U46619 was seen in vessels from Svep1+/- mice compared to littermate controls or when integrin α4β1 or α9β1 was inhibited. Inhibition studies suggested that this effect was mediated via VGCCs, PKC and Rho A/Rho kinase dependent mechanisms. CONCLUSIONS AND IMPLICATIONS Our studies reveal a novel role for SVEP1 and the integrins α4β1 and α9β1 in reducing VSMC contractility. This could provide an explanation for the genetic associations with blood pressure risk at the SVEP1 and ITGA9 loci.
Collapse
Affiliation(s)
- Gavin E. Morris
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Matthew J. Denniff
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Elisavet Karamanavi
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Sarah A. Andrews
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Renata B. Kostogrys
- Department of Human Nutrition, Faculty of Food TechnologyUniversity of Agriculture in KrakowKrakowPoland
| | - Vasiliki Bountziouka
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Maryam Ghaderi‐Najafabadi
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Noor Shamkhi
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - George McConnell
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Michael A. Kaiser
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | | | | | - Thorsten Kessler
- Department of Cardiology, German Heart Centre MunichTechnical University of MunichMunichGermany,German Centre of Cardiovascular Research (DZHK e. V.), Partner Site Munich Heart AllianceMunichGermany
| | - Richard D. Rainbow
- Department of Cardiovascular and Metabolic Medicine & Liverpool Centre for Cardiovascular ScienceUniversity of LiverpoolLiverpoolUK
| | - Nilesh J. Samani
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Thomas R. Webb
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| |
Collapse
|
36
|
Zhang L, Cunningham JW, Claggett BL, Jacob J, Mendelson MM, Serrano-Fernandez P, Kaiser S, Yates DP, Healey M, Chen CW, Turner GM, Patel-Murray NL, Zhao F, Beste MT, Laramie JM, Abraham WT, Jhund PS, Kober L, Packer M, Rouleau J, Zile MR, Prescott MF, Lefkowitz M, McMurray JJV, Solomon SD, Chutkow W. Aptamer Proteomics for Biomarker Discovery in Heart Failure With Reduced Ejection Fraction. Circulation 2022; 146:1411-1414. [PMID: 36029463 DOI: 10.1161/circulationaha.122.061481] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Luqing Zhang
- Novartis Institutes for Biomedical Research, Cambridge, MA (L.Z., J.J., M.M.M., D.P.Y., M.H., C.-W.C., G.M.T., N.L.P.-M., F.Z., M.T.B., J.M.L., W.C.)
| | | | - Brian L Claggett
- Brigham and Women's Hospital, Boston, MA (J.W.C., B.L.C., S.D.S.)
| | - Jaison Jacob
- Novartis Institutes for Biomedical Research, Cambridge, MA (L.Z., J.J., M.M.M., D.P.Y., M.H., C.-W.C., G.M.T., N.L.P.-M., F.Z., M.T.B., J.M.L., W.C.)
| | - Michael M Mendelson
- Novartis Institutes for Biomedical Research, Cambridge, MA (L.Z., J.J., M.M.M., D.P.Y., M.H., C.-W.C., G.M.T., N.L.P.-M., F.Z., M.T.B., J.M.L., W.C.)
| | | | - Sergio Kaiser
- Novartis Institutes for Biomedical Research, Basel, Switzerland (P.S.-F., S.K.)
| | - Denise P Yates
- Novartis Institutes for Biomedical Research, Cambridge, MA (L.Z., J.J., M.M.M., D.P.Y., M.H., C.-W.C., G.M.T., N.L.P.-M., F.Z., M.T.B., J.M.L., W.C.)
| | - Margaret Healey
- Novartis Institutes for Biomedical Research, Cambridge, MA (L.Z., J.J., M.M.M., D.P.Y., M.H., C.-W.C., G.M.T., N.L.P.-M., F.Z., M.T.B., J.M.L., W.C.)
| | - Chien-Wei Chen
- Novartis Institutes for Biomedical Research, Cambridge, MA (L.Z., J.J., M.M.M., D.P.Y., M.H., C.-W.C., G.M.T., N.L.P.-M., F.Z., M.T.B., J.M.L., W.C.)
| | - Gordon M Turner
- Novartis Institutes for Biomedical Research, Cambridge, MA (L.Z., J.J., M.M.M., D.P.Y., M.H., C.-W.C., G.M.T., N.L.P.-M., F.Z., M.T.B., J.M.L., W.C.)
| | - Natasha L Patel-Murray
- Novartis Institutes for Biomedical Research, Cambridge, MA (L.Z., J.J., M.M.M., D.P.Y., M.H., C.-W.C., G.M.T., N.L.P.-M., F.Z., M.T.B., J.M.L., W.C.)
| | - Faye Zhao
- Novartis Institutes for Biomedical Research, Cambridge, MA (L.Z., J.J., M.M.M., D.P.Y., M.H., C.-W.C., G.M.T., N.L.P.-M., F.Z., M.T.B., J.M.L., W.C.)
| | - Michael T Beste
- Novartis Institutes for Biomedical Research, Cambridge, MA (L.Z., J.J., M.M.M., D.P.Y., M.H., C.-W.C., G.M.T., N.L.P.-M., F.Z., M.T.B., J.M.L., W.C.)
| | - Jason M Laramie
- Novartis Institutes for Biomedical Research, Cambridge, MA (L.Z., J.J., M.M.M., D.P.Y., M.H., C.-W.C., G.M.T., N.L.P.-M., F.Z., M.T.B., J.M.L., W.C.)
| | | | - Pardeep S Jhund
- British Heart Foundation Cardiovascular Research Centre, University of Glasgow, United Kingdom (P.S.J., J.J.V.M.)
| | - Lars Kober
- Rigshospitalet Copenhagen University Hospital, Denmark (L.K.)
| | | | - Jean Rouleau
- Montreal Heart Institute and Université de Montréal, Canada (J.R.)
| | - Michael R Zile
- Ralph H. Johnson Department of Veterans Affairs Medical Center and Medical University of South Carolina, Charleston (M.R.Z.)
| | | | | | - John J V McMurray
- British Heart Foundation Cardiovascular Research Centre, University of Glasgow, United Kingdom (P.S.J., J.J.V.M.)
| | - Scott D Solomon
- Brigham and Women's Hospital, Boston, MA (J.W.C., B.L.C., S.D.S.)
| | - William Chutkow
- Novartis Institutes for Biomedical Research, Cambridge, MA (L.Z., J.J., M.M.M., D.P.Y., M.H., C.-W.C., G.M.T., N.L.P.-M., F.Z., M.T.B., J.M.L., W.C.)
| |
Collapse
|
37
|
Jung IH, Elenbaas JS, Burks KH, Amrute JM, Xiangyu Z, Alisio A, Stitziel NO. Vascular smooth muscle- and myeloid cell-derived integrin α9β1 does not directly mediate the development of atherosclerosis in mice. Atherosclerosis 2022; 360:15-20. [PMID: 36215801 PMCID: PMC9615102 DOI: 10.1016/j.atherosclerosis.2022.09.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/12/2022] [Accepted: 09/30/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND AIMS Sushi, von Willebrand factor type A, EGF pentraxin domain-containing 1 (SVEP1), an extracellular matrix protein, is a human coronary artery disease locus that promotes atherosclerosis. We previously demonstrated that SVEP1 induces vascular smooth muscle cell (VSMC) proliferation and an inflammatory phenotype in the arterial wall to enhance the development of atherosclerotic plaque. The only receptor known to interact with SVEP1 is integrin α9β1, a cell surface receptor that is expressed by VSMCs and myeloid lineage-derived monocytes and macrophages. Our previous in vitro studies suggested that integrin α9β1 was necessary for SVEP1-induced VSMC proliferation and inflammation; however, the underlying mechanisms mediated by integrin α9β1 in these cell types during the development of atherosclerosis remain poorly understood. METHODS AND RESULTS Here, using cell-specific gene targeting, we investigated the effects of the integrin α9β1 receptor on VSMCs and myeloid cells in mouse models of atherosclerosis. Interestingly, we found that depleting integrin α9β1 in either VSMCs or myeloid cells did not affect the formation or complexity of atherosclerotic plaque in vessels after either 8 or 16 weeks of high fat diet feeding. CONCLUSIONS Our results indicate that integrin α9β1 in these two cell types does not mediate the in vivo effect of SVEP1 in the development of atherosclerosis. Instead, our results suggest either the presence of other potential receptor(s) or alternative integrin α9β1-expressing cell types responsible for SVEP1 induced signaling in the development of atherosclerosis.
Collapse
Affiliation(s)
- In-Hyuk Jung
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Jared S Elenbaas
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA; Medical Scientist Training Program, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Kendall H Burks
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA; Medical Scientist Training Program, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Junedh M Amrute
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA; Medical Scientist Training Program, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Zhang Xiangyu
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Arturo Alisio
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Nathan O Stitziel
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA; McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, 63108, USA; Department of Genetics, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
| |
Collapse
|
38
|
Meng H, Ruan J, Yan Z, Chen Y, Liu J, Li X, Meng F. New Progress in Early Diagnosis of Atherosclerosis. Int J Mol Sci 2022; 23:8939. [PMID: 36012202 PMCID: PMC9409135 DOI: 10.3390/ijms23168939] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/30/2022] [Accepted: 08/06/2022] [Indexed: 11/18/2022] Open
Abstract
Coronary atherosclerosis is a potentially chronic circulatory condition that endangers human health. The biological cause underpinning cardiovascular disease is coronary atherosclerosis, and acute cardiovascular events can develop due to thrombosis, platelet aggregation, and unstable atherosclerotic plaque rupture. Coronary atherosclerosis is progressive, and three specific changes appear, with fat spots and stripes, atherosclerosis and thin-walled fiber atherosclerosis, and then complex changes in arteries. The progression and severity of cardiovascular disease are correlated with various levels of calcium accumulation in the coronary artery. The therapy and diagnosis of coronary atherosclerosis benefit from the initial assessment of the size and degree of calcification. This article will discuss the new progress in the early diagnosis of coronary atherosclerosis in terms of three aspects: imaging, gene and protein markers, and trace elements. This study intends to present the latest methods for diagnosing patients with early atherosclerosis through a literature review.
Collapse
Affiliation(s)
- Heyu Meng
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
| | - Jianjun Ruan
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
| | - Zhaohan Yan
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
| | - Yanqiu Chen
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
| | - Jinsha Liu
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
| | - Xiangdong Li
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
| | - Fanbo Meng
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
- Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute, Jilin University, Changchun 130033, China
| |
Collapse
|
39
|
Dynamic genome-wide gene expression and immune cell composition in the developing human placenta. J Reprod Immunol 2022; 151:103624. [DOI: 10.1016/j.jri.2022.103624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/23/2022] [Accepted: 04/13/2022] [Indexed: 11/23/2022]
|
40
|
Brandt KJ, Burger F, Baptista D, Roth A, Fernandes da Silva R, Montecucco F, Mach F, Miteva K. Single-Cell Analysis Uncovers Osteoblast Factor Growth Differentiation Factor 10 as Mediator of Vascular Smooth Muscle Cell Phenotypic Modulation Associated with Plaque Rupture in Human Carotid Artery Disease. Int J Mol Sci 2022; 23:1796. [PMID: 35163719 PMCID: PMC8836240 DOI: 10.3390/ijms23031796] [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] [Received: 12/19/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 12/18/2022] Open
Abstract
(1) Background: Vascular smooth muscle cells (VSMCs) undergo a complex phenotypic switch in response to atherosclerosis environmental triggers, contributing to atherosclerosis disease progression. However, the complex heterogeneity of VSMCs and how VSMC dedifferentiation affects human carotid artery disease (CAD) risk has not been clearly established. (2) Method: A single-cell RNA sequencing analysis of CD45- cells derived from the atherosclerotic aorta of Apolipoprotein E-deficient (Apoe-/-) mice on a normal cholesterol diet (NCD) or a high cholesterol diet (HCD), respecting the site-specific predisposition to atherosclerosis was performed. Growth Differentiation Factor 10 (GDF10) role in VSMCs phenotypic switch was investigated via flow cytometry, immunofluorescence in human atherosclerotic plaques. (3) Results: scRNAseq analysis revealed the transcriptomic profile of seven clusters, five of which showed disease-relevant gene signature of VSMC macrophagic calcific phenotype, VSMC mesenchymal chondrogenic phenotype, VSMC inflammatory and fibro-phenotype and VSMC inflammatory phenotype. Osteoblast factor GDF10 involved in ossification and osteoblast differentiation emerged as a hallmark of VSMCs undergoing phenotypic switch. Under hypercholesteremia, GDF10 triggered VSMC osteogenic switch in vitro. The abundance of GDF10 expressing osteogenic-like VSMCs cells was linked to the occurrence of carotid artery disease (CAD) events. (4) Conclusions: Taken together, these results provide evidence about GDF10-mediated VSMC osteogenic switch, with a likely detrimental role in atherosclerotic plaque stability.
Collapse
Affiliation(s)
- Karim J. Brandt
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Fabienne Burger
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Daniela Baptista
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Aline Roth
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Rafaela Fernandes da Silva
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 6627, Brazil
- Swiss Institute for Translational and Entrepreneurial Medicine, Freiburgstrasse 3, 3010 Bern, Switzerland
| | - Fabrizio Montecucco
- Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy;
- First Clinic of Internal Medicine, Department of Internal Medicine, Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| | - Francois Mach
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Kapka Miteva
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| |
Collapse
|