1
|
Mury P, Dagher O, Fortier A, Diaz A, Lamarche Y, Noly PE, Ibrahim M, Pagé P, Demers P, Bouchard D, Bernier PL, Poirier N, Moss E, Durrleman N, Jeanmart H, Pellerin M, Lettre G, Thorin-Trescases N, Carrier M, Thorin E. Quercetin Reduces Vascular Senescence and Inflammation in Symptomatic Male but Not Female Coronary Artery Disease Patients. Aging Cell 2025:e70108. [PMID: 40375481 DOI: 10.1111/acel.70108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/17/2025] [Accepted: 05/07/2025] [Indexed: 05/18/2025] Open
Abstract
Recent studies suggest that vascular senescence and its associated inflammation fuel the inflammaging to favor atherogenesis; whether these pathways can be therapeutically targeted in coronary artery disease (CAD) patients remains unknown. In a randomized, double-blind trial, 97 patients (78 men) undergoing coronary artery bypass graft surgery were treated with either quercetin (500 mg twice daily, 47 patients) or placebo (50 patients) for two days pre-surgery through hospital discharge. Primary outcomes were reduced inflammation and improved endothelial function ex vivo. Exploratory analyses included plasma proteomics and single-nuclei RNA sequencing of internal thoracic artery (ITA) samples. Quercetin treatment showed a trend toward reduced C-reactive protein at discharge (p = 0.073) and differentially modulated circulating inflammatory protein expression between men and women, with a pro-inflammatory effect of quercetin in females. Endothelial acetylcholine-induced relaxation improved significantly with quercetin (p = 0.049), with effects in men (p = 0.043) but not in women (p = 0.852). ITA transcriptomics revealed the overexpression of senescence and inflammaging pathways in male vascular cells, which quercetin reversed. In female cells, quercetin had minimal endothelial benefit and increased inflammaging in fibroblasts. In male cells, a candidate target of quercetin involves interactions between the receptor PLAUR and its ligands PLAU and SERPINE1. Post-operative atrial fibrillation incidence was significantly lower with quercetin, representing 4% of the patients compared to 18% in the placebo group (p = 0.033). In conclusion, short-term quercetin treatment effectively targeted vascular senescence in male CAD patients, improving inflammatory and functional outcomes. However, these benefits were not observed in female patients. Trial Registration: https://clinicaltrials.gov, NCT04907253.
Collapse
Affiliation(s)
- Pauline Mury
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Department of Pharmacology & Physiology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Olina Dagher
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Department of Pharmacology & Physiology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Calgary, Alberta, Canada
| | - Annik Fortier
- Department of Biostatistics, Montreal Health Innovations Coordinating Centre (MHICC), Montréal, Québec, Canada
| | - Ariel Diaz
- CIUSSS-MCQ, Université de Montréal, Campus Mauricie, Trois-Rivières, Québec, Canada
| | - Yoan Lamarche
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Pierre-Emmanuel Noly
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Marina Ibrahim
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Pierre Pagé
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Philippe Demers
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Denis Bouchard
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Pierre-Luc Bernier
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Nancy Poirier
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Emmanuel Moss
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Nicolas Durrleman
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Hughes Jeanmart
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Michel Pellerin
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Guillaume Lettre
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | | | - Michel Carrier
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Eric Thorin
- Montreal Heart Institute, Research Center, Université de Montréal, Montréal, Québec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
2
|
Faa G, Ziranu P, Pretta A, Cau F, Castagnola M, Spanu D, Saba G, D'Agata AP, Tiwari E, Suri JS, Scartozzi M, Saba L. Cancer-associated fibroblasts (CAFs) and plaque-associated fibroblasts (PAFs): Unraveling the cellular crossroads of atherosclerosis and cancer. Biomed Pharmacother 2025; 188:118145. [PMID: 40373629 DOI: 10.1016/j.biopha.2025.118145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 05/04/2025] [Accepted: 05/05/2025] [Indexed: 05/17/2025] Open
Abstract
Atherosclerosis is a complex process involving various cells and molecules within the atherosclerotic plaque. Recent evidence suggests that plaque-associated fibroblasts (PAFs), also known as atherosclerosis-associated fibroblasts (AAFs), might play a significant role in the development and progression of the disease. The microenvironment of the atherosclerotic plaque, resembling the tumor microenvironment (TME), includes various cellular populations like plaque-associated macrophages (PAMs), plaque-associated neutrophils (PANs), vascular smooth muscle cells (VSMCs), myeloid-derived suppressor cells (MDSCs), and PAFs. Similar to cancer-associated fibroblasts (CAFs) in tumors, PAFs exhibits a wide range of characteristics and functions. Their interactions with endothelial cells, smooth muscle cells, and other stromal cells, including adventitial fibroblast precursors, significantly influence atherosclerosis progression. Moreover, the ability of PAFs to express various markers such as alpha-SMA, Desmin, VEGF, and GFAP, highlights their diverse origins from different precursor cells, including vascular smooth muscle cells, endothelial cells, glial cells of the enteric nervous system, adventitial fibroblast precursors, as well as resident and circulating fibrocytes. This article explores the molecular interactions between PAFs, cells associated with atherosclerosis, and other stromal cells. It further examines the role of PAFs in the development and progression of atherosclerosis, and compares their features with those of CAFs. The research suggests that studying tumor-associated fibroblasts can help understand fibroblast subpopulations in atherosclerotic plaque. Identifying specific subpopulations could provide new insight into atherosclerosis complexity and lead to the development of innovative drugs for medical intervention.
Collapse
Affiliation(s)
- Gavino Faa
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy; Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | - Pina Ziranu
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy.
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Flaviana Cau
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Massimo Castagnola
- Laboratory of Proteomics, Centro Europeo di Ricerca sul Cervello, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Dario Spanu
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Giorgio Saba
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Alessandra Pia D'Agata
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Ekta Tiwari
- Department of Innovation. Global Biomedical Technologies, Inc., Roseville, CA 95661, USA
| | - Jasjit S Suri
- Department of ECE, Idaho State University, Pocatello, ID, 83209, USA; Department of CE, Graphics Era Deemed to be University, Dehradun 248002, India; University Center for Research & Development, Chandigarh University, Mohali, India; Symbiosis Institute of Technology, Nagpur Campus, Symbiosis International (Deemed University), Pune, INDIA; Stroke Diagnostic and Monitoring Division, AtheroPoint, Roseville, CA 95661, USA
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Luca Saba
- Department of Medical Sciences and Public Health, Unit of Radiology, University fo Cagliari, Cagliari, Italy
| |
Collapse
|
3
|
Yu B, Sopic M, Sluimer JC. Single-cell RNA sequencing (scRNA-seq) and its insights into cellular heterogeneity in atherosclerosis. Vascul Pharmacol 2025:107499. [PMID: 40345606 DOI: 10.1016/j.vph.2025.107499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/11/2025]
Abstract
Single-cell RNA sequencing (scRNA-seq) has transformed our understanding of cellular diversity in human biology, providing novel insights into disease mechanisms. In cardiovascular disease (CVD), scRNA-seq enables precise mapping of complex cell populations, uncovering unique cell types and states that influence disease progression and suggest new therapeutic targets. In atherosclerosis (AS), scRNA-seq has redefined plaque pathology by identifying distinct cell types, including endothelial cells (ECs), smooth muscle cells (SMCs), fibroblasts, macrophages, T cells, and B cells, each with specific roles in plaque stability, inflammation, and disease progression. In our review, we summarized these major cellular populations and their cellular heterogeneity in non-diseased and atherosclerotic aorta, as identified by scRNA-seq in mice and human tissues. We discussed conserved and species-specific subpopulations, their defining markers, and their functional implications in plaque progression. In addition, we integrated findings from scRNA-seq with experimental studies to highlight key molecular targets with therapeutic potential. In the future, these insights offer a refined cellular and molecular framework of atherosclerosis and may help the development of targeted interventions to promote plaque stabilization and reduce cardiovascular risk.
Collapse
Affiliation(s)
- Baixue Yu
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, 6229 HX Maastricht, the Netherlands.
| | - Miron Sopic
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg; Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia.
| | - Judith C Sluimer
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, 6229 HX Maastricht, the Netherlands; Aachen Maastricht Institute for CardioRenal research (AMICARE), 52074 Aachen, Germany; British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
4
|
Perry RN, Lenert G, Benavente ED, Ma A, Barbera N, Mokry M, de Kleijn DPV, de Winther MPJ, Mayr M, Björkegren JLM, den Ruijter HM, Civelek M. Female-biased vascular smooth muscle cell gene regulatory networks predict MYH9 as a key regulator of fibrous plaque phenotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645955. [PMID: 40236025 PMCID: PMC11996327 DOI: 10.1101/2025.03.28.645955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Atherosclerosis, a chronic inflammatory condition driving coronary artery disease (CAD), manifests in two primary plaque types: unstable atheromatous plaques and stable fibrous plaques. While significant research has focused on atheromatous plaques, recent studies emphasize the growing importance of fibrous plaques, particularly in females under 50 years of age, where erosion on fibrous plaques significantly contributes to coronary thrombosis. The molecular mechanisms underlying sex differences in atherosclerotic plaque characteristics, including vascular smooth muscle cell (VSMC) contributions, remain understudied. Therefore, we utilized sex-specific gene regulatory networks (GRNs) derived from VSMC gene expression data from 119 male and 32 female heart transplant donors to identify molecular drivers of fibrous plaques. GRN analysis revealed two female-biased networks in VSMC, GRN floralwhite and GRN yellowgreen , enriched for inflammatory signaling and actin remodeling pathways, respectively. Single-cell RNA sequencing of carotid plaques from female and male patients confirmed the sex specificity of these networks in VSMCs. Further sub cellular phenotyping of the single-cell RNA sequencing revealed a sex-specific gene expression signature within GRN yellowgreen for VSMCs enriched for contractile and vasculature development pathways. Bayesian network modeling of the GRN yellowgreen identified MYH9 as a key driver gene. Indeed, elevated MYH9 protein expression in atherosclerotic plaques was associated with higher smooth muscle cell content and lower lipid content in female plaques, suggesting its involvement in fibrous plaque formation. Further proteomic analysis confirmed MYH9's upregulation in female fibrous plaques only and its correlation with stable plaque features. These findings provide novel insights into sex-specific molecular mechanisms regulating fibrous plaque formation.
Collapse
|
5
|
Sukhavasi K, Mocci G, Ma L, Hodonsky CJ, Diez Benevante E, Muhl L, Liu J, Gustafsson S, Buyandelger B, Koplev S, Lendahl U, Vanlandewijck M, Singha P, Örd T, Beter M, Selvarajan I, Laakkonen JP, Väli M, den Ruijter HM, Civelek M, Hao K, Ruusalepp A, Betsholtz C, Järve H, Kovacic JC, Miller CL, Romanoski C, Kaikkonen MU, Björkegren JLM. Single-cell RNA sequencing reveals sex differences in the subcellular composition and associated gene-regulatory network activity of human carotid plaques. NATURE CARDIOVASCULAR RESEARCH 2025; 4:412-432. [PMID: 40211055 PMCID: PMC11994450 DOI: 10.1038/s44161-025-00628-y] [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: 02/23/2024] [Accepted: 02/17/2025] [Indexed: 04/12/2025]
Abstract
Carotid stenosis causes ischemic stroke in both sexes, but the clinical presentation and plaque characteristics differ. Here we run deep single-cell sequencing of 7,690 human carotid plaque cells from male and female patients. While we found no sex differences in major cell types, we identified a predominance of the osteogenic phenotype in smooth muscle cells, immunomodulating macrophages (MPs) and endothelial cells (ECs) undergoing endothelial-to-mesenchymal transition in females. In males, we found smooth muscle cells with the chondrocytic phenotype, MPs involved in tissue remodeling and ECs with angiogenic activity. Sex-biased subcellular clusters were integrated with tissue-specific gene-regulatory networks (GRNs) from the Stockholm-Tartu Atherosclerosis Reverse Network Engineering Task study. We identified GRN195 involved in angiogenesis and T cell-mediated cytotoxicity in male ECs, while in females, we found GRN33 and GRN122 related to TREM2-/TREM1+ MPs and endothelial-to-mesenchymal transition. The impact of GRN195 on EC proliferation in males was functionally validated, providing evidence for potential therapy targets for atherosclerosis that are sex specific.
Collapse
Grants
- 19TPA34910021 American Heart Association (American Heart Association, Inc.)
- R01 HL148167 NHLBI NIH HHS
- R01 HL148239 NHLBI NIH HHS
- PlaqOmics (18CVD02) Fondation Leducq
- R01 HG012773 NHGRI NIH HHS
- R01 HL168174 NHLBI NIH HHS
- AtheroGen (22CVD04) and PlaqOmics(18CVD02) Fondation Leducq
- R01 HL164577 NHLBI NIH HHS
- R01 HL166428 NHLBI NIH HHS
- research support from the NIH (R01HL148167, R01HG012773), New South Wales health grant RG194194, the Bourne Foundation, Snow Medical and Agilent
- Sydäntutkimussäätiö (Finnish Foundation for Cardiovascular Research)
- Sigrid Juséliuksen Säätiö (Sigrid Jusélius Foundation)
- Research Council of Finland, Competitive Funding to Strengthen University Research Profiles, 7th Call, profiling measure TransMed, (352968)
- The Research Council of Finland (328835), and GeneCellNano Flagship Program 337120
Collapse
Affiliation(s)
- Katyayani Sukhavasi
- Department of Cardiac Surgery and The Heart Clinic, Tartu University Hospital and Department of Cardiology, Institute of Clinical Medicine, Tartu University, Tartu, Estonia.
| | - Giuseppe Mocci
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Lijiang Ma
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chani J Hodonsky
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
| | - Ernest Diez Benevante
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Lars Muhl
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Jianping Liu
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Sonja Gustafsson
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Byambajav Buyandelger
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Simon Koplev
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Urban Lendahl
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Michael Vanlandewijck
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Prosanta Singha
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tiit Örd
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mustafa Beter
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Ilakya Selvarajan
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Johanna P Laakkonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Marika Väli
- Department of Pathological Anatomy and Forensic Sciences, Tartu University, Tartu, Estonia
| | - Hester M den Ruijter
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mete Civelek
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Arno Ruusalepp
- Department of Cardiac Surgery and The Heart Clinic, Tartu University Hospital and Department of Cardiology, Institute of Clinical Medicine, Tartu University, Tartu, Estonia
| | - Christer Betsholtz
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Heli Järve
- Department of Vascular Surgery and The Surgery Clinic, Tartu University Hospital, Tartu, Estonia
| | - Jason C Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical School, University of NSW, Sydney, New South Wales, Australia
| | - Clint L Miller
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Casey Romanoski
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Minna U Kaikkonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Johan L M Björkegren
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden.
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Clinical Gene Networks AB, Stockholm, Sweden.
| |
Collapse
|
6
|
Mury P, Cagnone G, Dagher O, Wünnemann F, Voghel G, Beaudoin M, Lambert M, Miquel G, Noly PE, Perrault LP, Carrier M, Thorin-Trescases N, Joyal JS, Lettre G, Thorin E. Senescence and Inflamm-Aging Are Associated With Endothelial Dysfunction in Men But Not Women With Atherosclerosis. JACC Basic Transl Sci 2024; 9:1163-1177. [PMID: 39534645 PMCID: PMC11551873 DOI: 10.1016/j.jacbts.2024.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 11/16/2024]
Abstract
Coronary artery disease (CAD) is more prevalent in men than in women, with endothelial dysfunction, prodromal to CAD, developing a decade earlier in middle-aged men. We investigated the molecular basis of this dimorphism ex vivo in arterial segments discarded during surgery of CAD patients. The results reveal a lower endothelial relaxant sensitivity in men, and a senescence-associated inflammaging transcriptomic signature in endothelial cells. In women, cellular metabolism and endothelial maintenance pathways are conserved. This suggests that senolytic therapies to reduce risk of cardiovascular events in women with CAD may not be as effective as in men.
Collapse
Affiliation(s)
- Pauline Mury
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Gael Cagnone
- University Hospital Sainte Justine Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Olina Dagher
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Calgary, Alberta, Canada
| | - Florian Wünnemann
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Guillaume Voghel
- Department of Family Medicine and Emergency Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Melissa Beaudoin
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Mélanie Lambert
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Géraldine Miquel
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Pierre-Emmanuel Noly
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Surgery, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Louis P. Perrault
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Surgery, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Michel Carrier
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Surgery, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | | | - Jean-Sébastien Joyal
- University Hospital Sainte Justine Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
- Department of Ophthalmology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Guillaume Lettre
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Eric Thorin
- Montreal Heart Institute Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Surgery, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Lan T, Palm KCA, Hoeben L, Diez Benavente E, Perry RN, Civelek M, de Kleijn DPV, den Ruijter HM, Pasterkamp G, Mokry M. Tobacco smoking is associated with sex- and plaque-type specific upregulation of CRLF1 in atherosclerotic lesions. Atherosclerosis 2024; 397:118554. [PMID: 39137621 DOI: 10.1016/j.atherosclerosis.2024.118554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 06/18/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND AND AIMS Tobacco smoking is a known risk factor for atherosclerotic disease, with more elevated risks in women compared to men. We hypothesized that atherosclerotic plaques from smokers show different gene expression patterns compared to non-smokers, in a sex-specific manner. METHODS Gene expression data of 625 carotid plaques (151 females and 474 males) were analyzed for differential gene expression between current smokers (n = 226) and non-smokers (n = 399). All analyses were stratified by sex and by molecular plaque characteristics. Finally, we projected the activity of gene regulatory networks and utilized single-cell transcriptomics from 38 plaques (26 males and 12 females) to interpret the sex- and plaque-type specific signals. RESULTS We observed higher expression levels of CRLF1 gene in atherosclerotic plaques from smokers compared to non-smokers (log2FC = 0.48, FDR = 0.012). CRLF1 upregulation was interacting with sex (p = 0.01) and was more pronounced in females (log2FC = 0.93, p = 1.53E-05) compared to males (log2FC = 0.35, p = 0.0018). Through single-cell RNA-seq analysis, we identified the highest CRLF1 expression within the transitioning and synthetic smooth muscle cell populations. CRLF1 expression was increased in fibro-inflammatory and fibro-cellular plaque types. Gene annotations pointed to increased expression of CRLF1 in networks with extracellular matrix related genes. CONCLUSIONS Atherosclerotic plaques from current smokers show sex-dependent upregulation of smooth muscle cell gene CRLF1. This may explain the different contributions of smoking to cardiovascular risk in females.
Collapse
Affiliation(s)
- Tian Lan
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands; Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Kaylin C A Palm
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Luka Hoeben
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Ernest Diez Benavente
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - R Noah Perry
- Center for Public Health Genomics, University of Virginia, Charlottesville, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, USA
| | - Mete Civelek
- Center for Public Health Genomics, University of Virginia, Charlottesville, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, USA
| | | | - Hester M den Ruijter
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Michal Mokry
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands; Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
8
|
Yu Y, Cai Y, Yang F, Yang Y, Cui Z, Shi D, Bai R. Vascular smooth muscle cell phenotypic switching in atherosclerosis. Heliyon 2024; 10:e37727. [PMID: 39309965 PMCID: PMC11416558 DOI: 10.1016/j.heliyon.2024.e37727] [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: 05/30/2024] [Revised: 08/25/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024] Open
Abstract
Atherosclerosis (AS) is a complex pathology process involving intricate interactions among various cells and biological processes. Vascular smooth muscle cells (VSMCs) are the predominant cell type in normal arteries, and under atherosclerotic stimuli, VSMCs respond to altered blood flow and microenvironment changes by downregulating contractile markers and switching their phenotype. This review overviews the diverse phenotypes of VSMCs, including the canonical contractile VSMCs, synthetic VSMCs, and phenotypes resembling macrophages, foam cells, myofibroblasts, osteoblasts/chondrocytes, and mesenchymal stem cells. We summarize their presumed protective and pro-atherosclerotic roles in AS development. Additionally, we underscore the molecular mechanisms and regulatory pathways governing VSMC phenotypic switching, encompassing transcriptional regulation, biochemical factors, plaque microenvironment, epigenetics, miRNAs, and the cytoskeleton, emphasizing their significance in AS development. Finally, we outline probable future research directions targeting VSMCs, offering insights into potential therapeutic strategies for AS management.
Collapse
Affiliation(s)
- Yanqiao Yu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yajie Cai
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| | - Furong Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| | - Yankai Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhuorui Cui
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Dazhuo Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| | - Ruina Bai
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| |
Collapse
|
9
|
Yin Y, Chen Y, Xu J, Liu B, Zhao Y, Tan X, Xiao M, Zhou Y, Zheng X, Xu Y, Han Z, Hu H, Li Z, Ou N, Lian W, Li Y, Su Z, Dai Y, Tang Y, Zhao L. Molecular and spatial signatures of human and rat corpus cavernosum physiopathological processes at single-cell resolution. Cell Rep 2024; 43:114760. [PMID: 39299236 DOI: 10.1016/j.celrep.2024.114760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/15/2024] [Accepted: 08/29/2024] [Indexed: 09/22/2024] Open
Abstract
The composition and cellular heterogeneity of the corpus cavernosum (CC) microenvironment have been characterized, but the spatial heterogeneity at the molecular level remains unexplored. In this study, we integrate single-cell RNA sequencing (scRNA-seq) and spatial transcriptome sequencing to comprehensively chart the spatial cellular landscape of the human and rat CC under normal and disease conditions. We observe differences in the proportions of cell subtypes and marker genes between humans and rats. Based on the analysis of the fibroblast (FB) niche, we also find that the enrichment scores of mechanical force signaling vary across different regions and correlate with the spatial distribution of FB subtypes. In vitro, the soft and hard extracellular matrix (ECM) induces the differentiation of FBs into apolipoprotein (APO)+ FBs and cartilage oligomeric matrix protein (COMP)+ FBs, respectively. In summary, our study provides a cross-species and physiopathological transcriptomic atlas of the CC, contributing to a further understanding of the molecular anatomy and regulation of penile erection.
Collapse
Affiliation(s)
- Yinghao Yin
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yuzhuo Chen
- Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Ultrasound, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Jiarong Xu
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Biao Liu
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yifan Zhao
- Department of Biostatistics & Informatics, Colorado School of Public Health, Aurora, CO, USA
| | - Xiaoli Tan
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Ming Xiao
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yihong Zhou
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Xiaoping Zheng
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yanghua Xu
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Zhitao Han
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Hongji Hu
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Zitaiyu Li
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Ningjing Ou
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Wenfei Lian
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yawei Li
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Zhongzhen Su
- Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Ultrasound, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yingbo Dai
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China.
| | - Yuxin Tang
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China.
| | - Liangyu Zhao
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China.
| |
Collapse
|
10
|
Zheng J, He J, Li H. FAM19A5 in vascular aging and osteoporosis: Mechanisms and the "calcification paradox". Ageing Res Rev 2024; 99:102361. [PMID: 38821416 DOI: 10.1016/j.arr.2024.102361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/05/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Aging induces a progressive decline in the vasculature's structure and function. Vascular aging is a determinant factor for vascular ailments in the elderly. FAM19A5, a recently identified adipokine, has demonstrated involvement in multiple vascular aging-related pathologies, including atherosclerosis, cardio-cerebral vascular diseases and cognitive deficits. This review summarizes the current understanding of FAM19A5' role and explores its putative regulatory mechanisms in various aging-related disorders, including cardiovascular diseases (CVDs), metabolic diseases, neurodegenerative diseases and malignancies. Importantly, we provide novel insights into the underlying therapeutic value of FAM19A5 in osteoporosis. Finally, we outline future perspectives on the diagnostic and therapeutic potential of FAM19A5 in vascular aging-related diseases.
Collapse
Affiliation(s)
- Jin Zheng
- Department of Geriatrics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Jieyu He
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Huahua Li
- Department of Geriatrics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China.
| |
Collapse
|
11
|
Diez Benavente E, Sakkers TR, van der Harst P, den Ruijter HM. Atherosclerotic plaque-specific methylation biomarkers in plasma cell-free DNA of female and male patients with coronary artery disease. Eur Heart J 2024; 45:2468-2470. [PMID: 38685672 DOI: 10.1093/eurheartj/ehae156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Affiliation(s)
- Ernest Diez Benavente
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Tim R Sakkers
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Pim van der Harst
- Department of Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Hester M den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
12
|
Diez Benavente E, Hartman RJG, Sakkers TR, Wesseling M, Sloots Y, Slenders L, Boltjes A, Mol BM, de Borst GJ, de Kleijn DPV, Prange KHM, de Winther MPJ, Kuiper J, Civelek M, van der Laan SW, Horvath S, Onland-Moret NC, Mokry M, Pasterkamp G, den Ruijter HM. Atherosclerotic Plaque Epigenetic Age Acceleration Predicts a Poor Prognosis and Is Associated With Endothelial-to-Mesenchymal Transition in Humans. Arterioscler Thromb Vasc Biol 2024; 44:1419-1431. [PMID: 38634280 DOI: 10.1161/atvbaha.123.320692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/25/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Epigenetic age estimators (clocks) are predictive of human mortality risk. However, it is not yet known whether the epigenetic age of atherosclerotic plaques is predictive for the risk of cardiovascular events. METHODS Whole-genome DNA methylation of human carotid atherosclerotic plaques (n=485) and of blood (n=93) from the Athero-Express endarterectomy cohort was used to calculate epigenetic age acceleration (EAA). EAA was linked to clinical characteristics, plaque histology, and future cardiovascular events (n=136). We studied whole-genome DNA methylation and bulk and single-cell transcriptomics to uncover molecular mechanisms of plaque EAA. We experimentally confirmed our in silico findings using in vitro experiments in primary human coronary endothelial cells. RESULTS Male and female patients with severe atherosclerosis had a median chronological age of 69 years. The median epigenetic age was 65 years in females (median EAA, -2.2 [interquartile range, -4.3 to 2.2] years) and 68 years in males (median EAA, -0.3 [interquartile range, -2.9 to 3.8] years). Patients with diabetes and a high body mass index had higher plaque EAA. Increased EAA of plaque predicted future events in a 3-year follow-up in a Cox regression model (univariate hazard ratio, 1.7; P=0.0034) and adjusted multivariate model (hazard ratio, 1.56; P=0.02). Plaque EAA predicted outcome independent of blood EAA (hazard ratio, 1.3; P=0.018) and of plaque hemorrhage (hazard ratio, 1.7; P=0.02). Single-cell RNA sequencing in plaque samples from 46 patients in the same cohort revealed smooth muscle and endothelial cells as important cell types in plaque EAA. Endothelial-to-mesenchymal transition was associated with EAA, which was experimentally confirmed by TGFβ-triggered endothelial-to-mesenchymal transition inducing rapid epigenetic aging in coronary endothelial cells. CONCLUSIONS Plaque EAA is a strong and independent marker of poor outcome in patients with severe atherosclerosis. Plaque EAA was linked to mesenchymal endothelial and smooth muscle cells. Endothelial-to-mesenchymal transition was associated with EAA, which was experimentally validated. Epigenetic aging mechanisms may provide new targets for treatments that reduce atherosclerosis complications.
Collapse
Affiliation(s)
- Ernest Diez Benavente
- Laboratory of Experimental Cardiology (E.D.B., R.J.G.H., T.R.S., Y.S., M.M., H.M.d.R.), University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Robin J G Hartman
- Laboratory of Experimental Cardiology (E.D.B., R.J.G.H., T.R.S., Y.S., M.M., H.M.d.R.), University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Tim R Sakkers
- Laboratory of Experimental Cardiology (E.D.B., R.J.G.H., T.R.S., Y.S., M.M., H.M.d.R.), University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Marian Wesseling
- Central Diagnostic Laboratory (M.W., L.S., A.B., S.W.v.d.L., M.M., G.P.), University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Yannicke Sloots
- Laboratory of Experimental Cardiology (E.D.B., R.J.G.H., T.R.S., Y.S., M.M., H.M.d.R.), University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Lotte Slenders
- Central Diagnostic Laboratory (M.W., L.S., A.B., S.W.v.d.L., M.M., G.P.), University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Arjan Boltjes
- Central Diagnostic Laboratory (M.W., L.S., A.B., S.W.v.d.L., M.M., G.P.), University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Barend M Mol
- Department of Vascular Surgery (B.M.M., G.J.d.B., D.P.V.d.K.), University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Gert J de Borst
- Department of Vascular Surgery (B.M.M., G.J.d.B., D.P.V.d.K.), University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Dominique P V de Kleijn
- Department of Vascular Surgery (B.M.M., G.J.d.B., D.P.V.d.K.), University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Koen H M Prange
- Division of Biotherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands (K.H.M.P., M.P.J.d.W., J.K.)
| | - Menno P J de Winther
- Division of Biotherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands (K.H.M.P., M.P.J.d.W., J.K.)
| | - Johan Kuiper
- Division of Biotherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands (K.H.M.P., M.P.J.d.W., J.K.)
| | - Mete Civelek
- Center for Public Health Genomics (M.C.), University of Virginia, Charlottesville
- Department of Biomedical Engineering (M.C.), University of Virginia, Charlottesville
| | - Sander W van der Laan
- Central Diagnostic Laboratory (M.W., L.S., A.B., S.W.v.d.L., M.M., G.P.), University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine (S.H.), University of California, Los Angeles
- Department of Biostatistics, Fielding School of Public Health (S.H.), University of California, Los Angeles
- Altos Labs, Cambridge Institute of Science, United Kingdom (S.H.)
| | - N Charlotte Onland-Moret
- Julius Center for Health Sciences and Primary Care (N.C.O.-M.), University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Michal Mokry
- Laboratory of Experimental Cardiology (E.D.B., R.J.G.H., T.R.S., Y.S., M.M., H.M.d.R.), University Medical Center Utrecht, Utrecht University, the Netherlands
- Central Diagnostic Laboratory (M.W., L.S., A.B., S.W.v.d.L., M.M., G.P.), University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Gerard Pasterkamp
- Central Diagnostic Laboratory (M.W., L.S., A.B., S.W.v.d.L., M.M., G.P.), University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Hester M den Ruijter
- Laboratory of Experimental Cardiology (E.D.B., R.J.G.H., T.R.S., Y.S., M.M., H.M.d.R.), University Medical Center Utrecht, Utrecht University, the Netherlands
| |
Collapse
|
13
|
Wu X, Zhang H. Omics Approaches Unveiling the Biology of Human Atherosclerotic Plaques. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:482-498. [PMID: 38280419 PMCID: PMC10988765 DOI: 10.1016/j.ajpath.2023.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 01/29/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall, characterized by the buildup of plaques with the accumulation and transformation of lipids, immune cells, vascular smooth muscle cells, and necrotic cell debris. Plaques with collagen-poor thin fibrous caps infiltrated by macrophages and lymphocytes are considered unstable because they are at the greatest risk of rupture and clinical events. However, the current histologic definition of plaque types may not fully capture the complex molecular nature of atherosclerotic plaque biology and the underlying mechanisms contributing to plaque progression, rupture, and erosion. The advances in omics technologies have changed the understanding of atherosclerosis plaque biology, offering new possibilities to improve risk prediction and discover novel therapeutic targets. Genomic studies have shed light on the genetic predisposition to atherosclerosis, and integrative genomic analyses expedite the translation of genomic discoveries. Transcriptomic, proteomic, metabolomic, and lipidomic studies have refined the understanding of the molecular signature of atherosclerotic plaques, aiding in data-driven hypothesis generation for mechanistic studies and offering new prospects for biomarker discovery. Furthermore, advancements in single-cell technologies and emerging spatial analysis techniques have unveiled the heterogeneity and plasticity of plaque cells. This review discusses key omics-based discoveries that have advanced the understanding of human atherosclerotic plaque biology, focusing on insights derived from omics profiling of human atherosclerotic vascular specimens.
Collapse
Affiliation(s)
- Xun Wu
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Hanrui Zhang
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York.
| |
Collapse
|
14
|
Pompei G, van de Hoef TP, den Ruijter HM, Kunadian V. Reading 2023 ESC ACS guidelines between the lines: Gaps in evidence in women. Eur J Clin Invest 2024; 54:e14148. [PMID: 38057949 DOI: 10.1111/eci.14148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/08/2023]
Affiliation(s)
- Graziella Pompei
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Tim P van de Hoef
- Division Heart and Lung, Department of Cardiology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Hester M den Ruijter
- Laboratory of Experimental Cardiology, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Vijay Kunadian
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Cardiothoracic Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|