1
|
Poppenborg T, Saljic A, Bruns F, Abu-Taha I, Dobrev D, Fender AC. A short history of the atrial NLRP3 inflammasome and its distinct role in atrial fibrillation. J Mol Cell Cardiol 2025; 202:13-23. [PMID: 40057301 DOI: 10.1016/j.yjmcc.2025.02.011] [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: 11/14/2024] [Revised: 01/21/2025] [Accepted: 02/24/2025] [Indexed: 04/23/2025]
Abstract
Inflammasomes are multiprotein complexes of the innate immune system that mediate inflammatory responses to infection and to local and systemic stress and tissue injury. The principal function is to facilitate caspase-1 auto-activation and subsequently maturation and release of the effectors interleukin (IL)-1β and IL-18. The atrial-specific NLRP3 inflammasome is a unifying causal feature of atrial fibrillation (AF) development, progression and recurrence after ablation. Many AF-associated risk factors and co-morbidities converge mechanistically on the activation of this central inflammatory signaling platform. This review presents the historical conceptual development of a distinct atrial inflammasome and its potential causal involvement in AF. We follow the early observations linking systemic and local inflammation with AF, to the emergence of an atrial-intrinsic NLRP3 inflammasome operating within not just immune cells but also in resident atrial fibroblasts and cardiomyocytes. We outline the key developments in understanding how the atrial NLRP3 inflammasome and its effector IL-1β contribute causally to cellular and tissue-level arrhythmogenesis in different pathological settings, and outline candidate therapeutic concepts verified in preclinical models of atrial cardiomyopathy and AF.
Collapse
Affiliation(s)
| | - Arnela Saljic
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Florian Bruns
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany
| | - Issam Abu-Taha
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA; Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada
| | - Anke C Fender
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
2
|
Yuan Y, Martsch P, Chen X, Martinez E, Li L, Song J, Poppenborg T, Bruns F, Kim JH, Kamler M, Martin JF, Abu-Taha I, Dobrev D, Li N. Atrial cardiomyocyte-restricted cleavage of gasdermin D promotes atrial arrhythmogenesis. Eur Heart J 2025; 46:1250-1262. [PMID: 39927987 PMCID: PMC11959185 DOI: 10.1093/eurheartj/ehaf024] [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: 04/16/2024] [Revised: 08/21/2024] [Accepted: 01/14/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND AIMS Enhanced inflammatory signalling causally contributes to atrial fibrillation (AF) development. Gasdermin D (GSDMD) is an important downstream effector of several inflammasome pathways. However, the role of GSDMD, particularly the cleaved N-terminal (NT)-GSDMD, in non-immune cells remains elusive. This study aimed to elucidate the function of NT-GSDMD in atrial cardiomyocytes (ACMs) and determine its contribution to atrial arrhythmogenesis. METHODS Human atrial appendages were used to assess the protein levels and localization. A modified adeno-associated virus 9 was employed to establish ACM-restricted overexpression of NT-GSDMD in mice. RESULTS The cleavage of GSDMD was enhanced in ACMs of AF patients. Atrial cardiomyocyte-restricted overexpression of NT-GSDMD in mice increased susceptibility to pacing-induced AF. The NT-GSDMD pore formation facilitated interleukin-1β secretion from ACMs, promoting macrophage infiltration, while up-regulating 'endosomal sorting complexes required for transport'-mediated membrane-repair mechanisms, which prevented inflammatory cell death (pyroptosis) in ACMs. Up-regulated NT-GSDMD directly targeted mitochondria, increasing mitochondrial reactive oxygen species (ROS) generation, which triggered proarrhythmic calcium-release events. The NT-GSDMD-induced arrhythmogenesis was mitigated by the mitochondrial-specific antioxidant MitoTEMPO. A mutant NT-GSDMD lacking pore-formation capability failed to cause mitochondrial dysfunction or induce atrial arrhythmia. Genetic ablation of Gsdmd prevented spontaneous AF development in a mouse model. CONCLUSIONS These findings establish a unique pyroptosis-independent role of NT-GSDMD in ACMs and arrhythmogenesis, which involves ROS-driven mitochondrial dysfunction. Mitochondrial-targeted therapy, either by reducing ROS production or inhibition of GSDMD, prevents AF inducibility, positioning GSDMD as a novel therapeutic target for AF prevention.
Collapse
Affiliation(s)
- Yue Yuan
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, One Baylor Plaza, MS: BCM285, Houston, TX 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Pascal Martsch
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Xiaohui Chen
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, One Baylor Plaza, MS: BCM285, Houston, TX 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Enrique Martinez
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, One Baylor Plaza, MS: BCM285, Houston, TX 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Luge Li
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, One Baylor Plaza, MS: BCM285, Houston, TX 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Jia Song
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, One Baylor Plaza, MS: BCM285, Houston, TX 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Theresa Poppenborg
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Florian Bruns
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Jong Hwan Kim
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Cardiomyocyte Renewal Laboratory, The Texas Heart Institute, Houston, TX, USA
| | - Markus Kamler
- Department of Thoracic and Cardiovascular Surgery, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - James F Martin
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Cardiomyocyte Renewal Laboratory, The Texas Heart Institute, Houston, TX, USA
| | - Issam Abu-Taha
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Na Li
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, One Baylor Plaza, MS: BCM285, Houston, TX 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
3
|
Beaudrey T, Bedo D, Weschler C, Caillard S, Florens N. From Risk Assessment to Management: Cardiovascular Complications in Pre- and Post-Kidney Transplant Recipients: A Narrative Review. Diagnostics (Basel) 2025; 15:802. [PMID: 40218153 PMCID: PMC11988545 DOI: 10.3390/diagnostics15070802] [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: 02/14/2025] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/14/2025] Open
Abstract
Kidney transplantation remains the best treatment for chronic kidney failure, offering better outcomes and quality of life compared with dialysis. Cardiovascular disease (CVD) is a major cause of morbidity and mortality in kidney transplant recipients and is associated with decreased patient survival and worse graft outcomes. Post-transplant CVD results from a complex interaction between traditional cardiovascular risk factors, such as hypertension and diabetes, and risk factors specific to kidney transplant recipients including chronic kidney disease, immunosuppressive drugs, or vascular access. An accurate assessment of cardiovascular risk is now needed to optimize the management of cardiovascular comorbidities through the detection of risk factors and the screening of hidden pretransplant coronary artery disease. Promising new strategies are emerging, such as GLP-1 receptor agonists and SGLT2 inhibitors, with a high potential to mitigate cardiovascular complications, although further research is needed to determine their role in kidney transplant recipients. Despite this progress, a significant gap remains in understanding the optimal management of post-transplant CVD, especially coronary artery disease, stroke, and peripheral artery disease. Addressing these challenges is essential to improve the short- and long-term outcomes in kidney transplant recipients. This narrative review aims to provide a comprehensive overview of cardiovascular risk assessment and post-transplant CVD management.
Collapse
Affiliation(s)
- Thomas Beaudrey
- Nephrology Department, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France; (T.B.); (D.B.); (C.W.); (S.C.)
- Inserm UMR_S 1109 Immuno-Rhumatology Laboratory, Translational Medicine Federation of Strasbourg (FMTS), FHU Target, Faculté de Médecine, Université de Strasbourg, 67000 Strasbourg, France
| | - Dimitri Bedo
- Nephrology Department, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France; (T.B.); (D.B.); (C.W.); (S.C.)
- Inserm UMR_S 1109 Immuno-Rhumatology Laboratory, Translational Medicine Federation of Strasbourg (FMTS), FHU Target, Faculté de Médecine, Université de Strasbourg, 67000 Strasbourg, France
| | - Célia Weschler
- Nephrology Department, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France; (T.B.); (D.B.); (C.W.); (S.C.)
| | - Sophie Caillard
- Nephrology Department, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France; (T.B.); (D.B.); (C.W.); (S.C.)
- Inserm UMR_S 1109 Immuno-Rhumatology Laboratory, Translational Medicine Federation of Strasbourg (FMTS), FHU Target, Faculté de Médecine, Université de Strasbourg, 67000 Strasbourg, France
| | - Nans Florens
- Nephrology Department, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France; (T.B.); (D.B.); (C.W.); (S.C.)
- Inserm UMR_S 1109 Immuno-Rhumatology Laboratory, Translational Medicine Federation of Strasbourg (FMTS), FHU Target, Faculté de Médecine, Université de Strasbourg, 67000 Strasbourg, France
- INI-CRCT (Cardiovascular and Renal Trialists), F-CRIN Network, 67000 Strasbourg, France
| |
Collapse
|
4
|
Nattel S. Emerging themes in the molecular pathophysiology of atrial fibrillation. Heart Rhythm 2025; 22:876-877. [PMID: 40023521 DOI: 10.1016/j.hrthm.2024.12.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 12/27/2024] [Indexed: 03/04/2025]
Affiliation(s)
- Stanley Nattel
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada; Research Center and Department of Medicine, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
5
|
Moreno-Loaiza O, Soares VC, de Assumpção Souza M, Vera-Nuñez N, Rodriguez de Yurre Guirao A, da Silva TP, Pozes AB, Perticarrari L, Monteiro E, Albino MC, Silva SB, Dias SSG, Maciel L, Muzi-Filho H, de Oliveira DF, Braga BC, Diniz LP, Cruz MC, Barbosa SR, Castro-Junior AB, Conde L, Cabral-Castro MJ, de Souza OF, Tavares Pinheiro MV, Araújo de Oliveira Junior N, Rezende de Siqueira L, Cosenza RP, Munhoz da Fontoura C, Secco JCP, da Rocha Ferreira J, Silvestre de Sousa A, Albuquerque D, Luiz RR, Nicolau-Neto P, Pretti MA, Boroni M, Bonamino MH, Kasai-Brunswick TH, Mello DB, Gonçalves-Silva T, Ramos IP, Bozza FA, Madeiro JPDV, Pedrosa RC, Carneiro-Ramos MS, da Silva Martinho H, Bozza PT, Mesquita de Souza F, Victor Lucena da Silva G, Cunha TM, Uzelac I, Fenton F, Moll-Bernardes R, Paiva CN, Escobar AL, Medei E. IL-1β enhances susceptibility to atrial fibrillation in mice by acting through resident macrophages and promoting caspase-1 expression. NATURE CARDIOVASCULAR RESEARCH 2025; 4:312-329. [PMID: 39915330 PMCID: PMC11980030 DOI: 10.1038/s44161-025-00610-8] [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: 05/09/2023] [Accepted: 01/09/2025] [Indexed: 03/19/2025]
Abstract
Atrial fibrillation (AF) is more prevalent in patients with elevated interleukin (IL)-1β levels. Here we show that daily administration of IL-1β for 15 days sensitizes mice to AF, leading to fibrosis, accumulation of β-pleated sheet proteins in the left atrium, and systemic inflammation, resembling the pathophysiological changes observed in patients with AF. IL-1β administration creates a positive feedback loop, dependent on the IL-1 receptor (IL-1R) activity in cardiac resident macrophages. This results in increased caspase-1 maturation in the left atrium and elevated Il1b and Casp1 transcription in atrial macrophages. IL-1β treatment accelerated action potential and Ca2+ restitution in the left atrium, leading to action-potential shortening. This, along with increased caspase-1 maturation and IL-1R signaling, was essential for inducing AF. Lack of IL-1R in macrophages, but not cardiomyocytes, prevented IL-1β-induced AF sensitivity. By depleting recruited macrophages or deleting IL-1R specifically in cardiac resident macrophages, we further demonstrate that IL-1β/IL-1R signaling in these resident macrophages is responsible for increased AF susceptibility. These findings offer insights into the therapeutic potential of targeting IL-1β/IL-1R signaling in patients with AF and emphasize the importance of recognizing different underlying causes in this patient group.
Collapse
Affiliation(s)
- Oscar Moreno-Loaiza
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Vinicius Cardoso Soares
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, Brazil
- Program of Immunology and Inflammation, Federal University of Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Manuela de Assumpção Souza
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Narendra Vera-Nuñez
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Tatiana Pereira da Silva
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Beatriz Pozes
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Larissa Perticarrari
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Evelin Monteiro
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria Clara Albino
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sophia Barros Silva
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Suelen Silva Gomes Dias
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, Brazil
| | - Leonardo Maciel
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Humberto Muzi-Filho
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Dahienne Ferreira de Oliveira
- Institute of Medical Biochemistry Leopoldo de Meis, Rio de Janeiro, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno Cabral Braga
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luan Pereira Diniz
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mario Costa Cruz
- Centro de Facilidades e Apoio à Pesquisa (CEFAP), Universidade de São Paulo (USP), São Paulo, Brazil
| | | | | | - Luciana Conde
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mauro Jorge Cabral-Castro
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Pathology Department, Fluminense Federal University, Niterói, Brazil
| | | | | | | | | | | | | | | | | | | | | | - Ronir Raggio Luiz
- Institute for Studies in Public Health-IESC, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Clementino Fraga University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro Nicolau-Neto
- Molecular Carcinogenesis Program, Research Coordination, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Marco Antonio Pretti
- Cell and Gene Therapy Program, Research Coordination, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Mariana Boroni
- Laboratory of Bioinformatics and Computational Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Martin Hernán Bonamino
- Molecular Carcinogenesis Program, Research Coordination, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
- Vice-Presidency of Research and Biological Collections (VPPCB), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Tais Hanae Kasai-Brunswick
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Debora Bastos Mello
- National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Triciana Gonçalves-Silva
- National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Isalira Peroba Ramos
- National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernando A Bozza
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
- Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | | | - Roberto Coury Pedrosa
- Clementino Fraga University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Patrícia T Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, Brazil
| | - Fernanda Mesquita de Souza
- Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, Ribeirão Preto, Brazil
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Gabriel Victor Lucena da Silva
- Center for Natural and Human Sciences, Federal University of ABC, Santo André, Brazil
- Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, Ribeirão Preto, Brazil
| | - Thiago M Cunha
- Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, Ribeirão Preto, Brazil
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Ilija Uzelac
- Virginia Commonwealth University, Richmond, VA, USA
| | | | | | - Claudia N Paiva
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ariel L Escobar
- Department of Bioengineering, School of Engineering, University of California, Merced, CA, USA
| | - Emiliano Medei
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil.
- National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
6
|
Keefe JA, Wang J, Song J, Ni L, Wehrens X. Immune cells and arrhythmias. Cardiovasc Res 2025:cvaf017. [PMID: 39937651 DOI: 10.1093/cvr/cvaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/12/2024] [Accepted: 01/07/2025] [Indexed: 02/14/2025] Open
Abstract
Cardiac arrhythmias are a significant cause of morbidity and mortality worldwide. Emerging evidence has demonstrated that resident and infiltrating cardiac immune cells play direct, mechanistic roles in arrhythmia onset and progression. In this review, we provide a comprehensive summary and expert commentary on the role of each immune cell subtype in the pathogenesis of atrial and ventricular arrhythmias.
Collapse
Affiliation(s)
- Joshua A Keefe
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jian Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- Shenzen Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzen, China
| | - Li Ni
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Xander Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Space Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
7
|
Goette A. Atrial fibrillation does not equal atrial fibrillation: The important prognostic implications of new-onset atrial fibrillation. IJC HEART & VASCULATURE 2025; 56:101572. [PMID: 39720340 PMCID: PMC11667165 DOI: 10.1016/j.ijcha.2024.101572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Affiliation(s)
- Andreas Goette
- Corresponding author at: St. Vincenz-Hospital Paderborn, Dept. of Cardiology and Intensive Care Medicine, Am Busdorf 2, 33098 Paderborn, Germany.
| |
Collapse
|
8
|
Andonian BJ, Hippensteel JA, Abuabara K, Boyle EM, Colbert JF, Devinney MJ, Faye AS, Kochar B, Lee J, Litke R, Nair D, Sattui SE, Sheshadri A, Sherman AN, Singh N, Zhang Y, LaHue SC. Inflammation and aging-related disease: A transdisciplinary inflammaging framework. GeroScience 2025; 47:515-542. [PMID: 39352664 PMCID: PMC11872841 DOI: 10.1007/s11357-024-01364-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
Inflammaging, a state of chronic, progressive low-grade inflammation during aging, is associated with several adverse clinical outcomes, including frailty, disability, and death. Chronic inflammation is a hallmark of aging and is linked to the pathogenesis of many aging-related diseases. Anti-inflammatory therapies are also increasingly being studied as potential anti-aging treatments, and clinical trials have shown benefits in selected aging-related diseases. Despite promising advances, significant gaps remain in defining, measuring, treating, and integrating inflammaging into clinical geroscience research. The Clin-STAR Inflammation Research Interest Group was formed by a group of transdisciplinary clinician-scientists with the goal of advancing inflammaging-related clinical research and improving patient-centered care for older adults. Here, we integrate insights from nine medical subspecialties to illustrate the widespread impact of inflammaging on diseases linked to aging, highlighting the extensive opportunities for targeted interventions. We then propose a transdisciplinary approach to enhance understanding and treatment of inflammaging that aims to improve comprehensive care for our aging patients.
Collapse
Affiliation(s)
- Brian J Andonian
- Division of Rheumatology and Immunology, Duke University School of Medicine, Durham, NC, USA.
| | - Joseph A Hippensteel
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katrina Abuabara
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Eileen M Boyle
- Department of Haematology, University College London Cancer Institute, London, UK
| | - James F Colbert
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael J Devinney
- Division of Critical Care, Department of Anesthesiology, Duke University School of Medicine, Durham, NC, USA
| | - Adam S Faye
- Division of Gastroenterology, Department of Population Health, NYU Langone Medical Center, New York, NY, USA
| | - Bharati Kochar
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Jiha Lee
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Rachel Litke
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Devika Nair
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sebastian E Sattui
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anoop Sheshadri
- Division of Nephrology, Department of Medicine, University of California, San Francisco, Nephrology Section, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | | | - Namrata Singh
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Yinan Zhang
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sara C LaHue
- Department of Neurology, School of Medicine, and the UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
9
|
Li Z, Wu H, Yao F, Li Y, Li Y, Xie SA, Yu F, Fu Y, Wang L, Zhou J, Kong W. Runx2-NLRP3 axis orchestrates matrix stiffness-evoked vascular smooth muscle cell inflammation. Am J Physiol Cell Physiol 2025; 328:C467-C482. [PMID: 39761974 DOI: 10.1152/ajpcell.00448.2024] [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: 07/02/2024] [Revised: 12/22/2024] [Accepted: 12/22/2024] [Indexed: 01/24/2025]
Abstract
Arterial stiffening is a hallmark of chronic kidney disease (CKD)-related cardiovascular events and is primarily attributed to the elevated matrix stiffness. Stiffened arteries are accompanied by low-grade inflammation, but the causal effects of matrix stiffness on inflammation remain unknown. For analysis of the relationship between arterial stiffness and vascular inflammation, pulse-wave velocity (PWV) and aortic inflammatory markers were analyzed in an adenine-induced mouse model of CKD in chronological order. Compared with their control littermates, mice with CKD showed elevated arterial stiffness at the early stage of disease progression, which preceded the onset of vascular inflammation. Correspondingly, the increase of matrix stiffness induced vascular smooth muscle cells (VSMCs) to transdifferentiate into an inflammatory phenotype, as indicated by the increased expression and secretion of MCP-1, IL-6, IL-1β, and IL-18. RNA-sequencing analysis of stiff matrix-cultured VSMCs and bioinformatics analysis with the ChIP-Atlas database revealed the potential involvement of the transcription factor Runx2. The expression and the nuclear localization of Runx2 were significantly increased in stiff matrix-cultured VSMCs. High-throughput ChIP-sequencing and promoter luciferase assays further revealed that NLRP3 was directly transcriptionally regulated by Runx2. The inhibition of Runx2 or NLRP3 inflammasome abrogated the proinflammatory effect of matrix stiffening on VSMCs. Together, these data revealed that arterial stiffness precedes vascular inflammatory responses in CKD mice and that the Runx2-NLRP3 axis orchestrates matrix stiffness and the VSMC inflammatory phenotype, which may contribute to the pathogenic role in arterial stiffness-related vascular inflammation and CKD-related cardiovascular complications.NEW & NOTEWORTHY As a hallmark of chronic kidney disease (CKD), arterial stiffening is related to increased vascular inflammation and cardiovascular morbidity, whereas the underlying mechanism is unclear. The study demonstrates that increased arterial stiffness precedes the onset of vascular inflammation, and matrix stiffness stimulates the transdifferentiation of vascular smooth muscle cells (VSMCs) to an inflammatory phenotype via activating Runx2-NLRP3 signaling, which provides novel insights into CKD-related cardiovascular disorder treatment.
Collapse
MESH Headings
- Animals
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Vascular Stiffness/physiology
- Core Binding Factor Alpha 1 Subunit/metabolism
- Core Binding Factor Alpha 1 Subunit/genetics
- Mice
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Inflammation/metabolism
- Inflammation/pathology
- Male
- Mice, Inbred C57BL
- Signal Transduction
- Cells, Cultured
- Disease Models, Animal
- Aorta/metabolism
- Aorta/pathology
Collapse
Affiliation(s)
- Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Hao Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Fang Yao
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China
| | - Yiran Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yanjie Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Si-An Xie
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Fang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Li Wang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
10
|
Liu Y, Liu H, Sun D, Zheng Y, Tse G, Chen K, Qiu J, Wu S, Liu T. Association of Estimated Glomerular Filtration Rate (eGFR) and High-Sensitivity C-Reactive Protein (Hs-CRP) with the Risk of New-Onset Atrial Fibrillation in Patients with Diabetes. J Inflamm Res 2025; 18:91-103. [PMID: 39780989 PMCID: PMC11708200 DOI: 10.2147/jir.s493068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
Background Both renal function decline and systemic inflammation may synergistically increase the risk of atrial fibrillation (AF). This study investigates the association between estimated glomerular filtration rate (eGFR) and high-sensitivity C-reactive protein (hs-CRP) levels with the risk of new-onset AF in patients with diabetes mellitus. Methods We included diabetic patients without AF who participated in physical exams in the Kailuan Study from 2006 to 2010. Participants were categorized into four groups based on baseline eGFR and hs-CRP levels: 1) high eGFR (≥60 mL/min/1.73m²) and low hs-CRP (<3 mg/L) (n=6,915), 2) high eGFR and high hs-CRP (≥3 mg/L) (n=3,154), 3) low eGFR (<60 mL/min/1.73m²) and low hs-CRP (n=4,638), 4) low eGFR and high hs-CRP (n=1,809). We employed multivariable Cox regression analysis to evaluate the relationships between eGFR, hs-CRP, and new-onset AF, adjusting for confounders including smoking status, alcohol consumption, blood pressure, fasting blood glucose (FBG), heart rate, lipid levels, body mass index (BMI), and medication usage. Competing risk analysis was also performed. Results Among 16,516 patients, 222 developed new-onset AF over a mean follow-up of 12.6 years. After adjusting for confounders, elevated hs-CRP and reduced eGFR were significantly associated with higher risk of new-onset AF compared to the high eGFR/low hs-CRP group. These findings remained consistent after excluding AF cases within the first 2-year. No significant interaction between eGFR and hs-CRP was observed (P=0.227). Subgroup analysis revealed that the combination of eGFR and hs-CRP had predictive value primarily in males under 60 years of age, individuals with FBG <9 mmol/L, hypertension, and those not on hypoglycemic medications. Conclusion In diabetic patients, decreased eGFR and elevated hs-CRP were independently linked to an increased risk of new-onset AF, emphasizing the importance of monitoring these factors for early detection and prevention of AF.
Collapse
Affiliation(s)
- Ying Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Hongmin Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
- Department of Cardiology, Kailuan General Hospital, Tangshan, 063001, People’s Republic of China
| | - Dongkun Sun
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Yi Zheng
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
- School of Nursing and Health Sciences, Hong Kong Metropolitan University, Hong Kong, People’s Republic of China
- Diabetes Research Unit, Cardiovascular Analytics Group, PowerHealth Research Institute, Hong Kong, People’s Republic of China
| | - Kangyin Chen
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Jiuchun Qiu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital, Tangshan, 063001, People’s Republic of China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| |
Collapse
|
11
|
Saljic A, Heijman J, Dobrev D. From Atrial Small-conductance Calcium-activated Potassium Channels to New Antiarrhythmics. Eur Cardiol 2024; 19:e26. [PMID: 39872420 PMCID: PMC11770539 DOI: 10.15420/ecr.2024.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/27/2024] [Indexed: 01/30/2025] Open
Abstract
Despite significant advances in its management, AF remains a major healthcare burden affecting millions of individuals. Rhythm control with antiarrhythmic drugs or catheter ablation has been shown to improve symptoms and outcomes in AF patients, but current treatment options have limited efficacy and/or significant side-effects. Novel mechanism-based approaches could potentially be more effective, enabling improved therapeutic strategies for managing AF. Small-conductance calcium-activated potassium (SK or KCa2.x) channels encoded by KCNN1-3 have recently gathered interest as novel antiarrhythmic targets with potential atrial-predominant effects. Here, the molecular composition of smallconductance calcium-activated potassium channels and their complex regulation in AF as the basis for understanding the distinct mechanism of action of pore-blockers (apamin, UCL1684, ICAGEN) and modulators of calcium-dependent activation (NS8593, AP14145, AP30663) are summarised. Furthermore, the preclinical and early clinical evidence for the role of small-conductance calcium-activated potassium channel inhibitors in the treatment of AF are reviewed.
Collapse
Affiliation(s)
- Arnela Saljic
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagen, Denmark
- Institute of Pharmacology, West German Heart and Vascular Center, University of Duisburg-EssenEssen, Germany
| | - Jordi Heijman
- Gottfried Schatz Research Centre, Division of Medical Physics & Biophysics, Medical University of GrazGraz, Austria
- Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Maastricht UniversityMaastricht, the Netherlands
| | - Dobromir Dobrev
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagen, Denmark
- Departments of Medicine and Research Centre, Montreal Heart Institute and Université de MontréalMontreal, Canada
- Department of Integrative Physiology, Baylor College of MedicineHouston, TX, US
| |
Collapse
|
12
|
Beikoghli Kalkhoran S, Basalay M, He Z, Golforoush P, Roper T, Caplin B, Salama AD, Davidson SM, Yellon DM. Investigating the cause of cardiovascular dysfunction in chronic kidney disease: capillary rarefaction and inflammation may contribute to detrimental cardiovascular outcomes. Basic Res Cardiol 2024; 119:937-955. [PMID: 39472324 PMCID: PMC11628583 DOI: 10.1007/s00395-024-01086-6] [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: 06/27/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 12/10/2024]
Abstract
Myocardial ischemia-reperfusion (IR) injury is a major cause of morbidity and mortality in patients with chronic kidney disease (CKD). The most frequently used and representative experimental model is the rat dietary adenine-induced CKD, which leads to CKD-associated CVD. However, the continued intake of adenine is a potential confounding factor. This study investigated cardiovascular dysfunction following brief adenine exposure, CKD development and return to a normal diet. Male Wistar rats received a 0.3% adenine diet for 10 weeks and normal chow for an additional 8 weeks. Kidney function was assessed by urinalysis and histology. Heart function was assessed by echocardiography. Sensitivity to myocardial IR injury was assessed using the isolated perfused rat heart (Langendorff) model. The inflammation profile of rats with CKD was assessed via cytokine ELISA, tissue histology and RNA sequencing. Induction of CKD was confirmed by a significant increase in plasma creatinine and albuminuria. Histology revealed extensive glomerular and tubular damage. Diastolic dysfunction, measured by the reduction of the E/A ratio, was apparent in rats with CKD even following a normal diet. Hearts from rats with CKD had significantly larger infarcts after IR injury. The CKD rats also had statistically higher levels of markers of inflammation including myeloperoxidase, KIM-1 and interleukin-33. RNA sequencing revealed several changes including an increase in inflammatory signaling pathways. In addition, we noted that CKD induced significant cardiac capillary rarefaction. We have established a modified model of adenine-induced CKD, which leads to cardiovascular dysfunction in the absence of adenine. Our observations of capillary rarefaction and inflammation suggest that these may contribute to detrimental cardiovascular outcomes.
Collapse
Affiliation(s)
- Siavash Beikoghli Kalkhoran
- The Hatter Cardiovascular Institute, University College London & UCL Hospital, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Maryna Basalay
- The Hatter Cardiovascular Institute, University College London & UCL Hospital, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Zhenhe He
- The Hatter Cardiovascular Institute, University College London & UCL Hospital, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Pelin Golforoush
- The Hatter Cardiovascular Institute, University College London & UCL Hospital, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Tayeba Roper
- Centre for Kidney and Bladder Health, Royal Free Hospital, University College London, London, England, UK
| | - Ben Caplin
- Centre for Kidney and Bladder Health, Royal Free Hospital, University College London, London, England, UK
| | - Alan D Salama
- Centre for Kidney and Bladder Health, Royal Free Hospital, University College London, London, England, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London & UCL Hospital, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London & UCL Hospital, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
13
|
Niskala A, Heijman J, Dobrev D, Jespersen T, Saljic A. Targeting the NLRP3 inflammasome signalling for the management of atrial fibrillation. Br J Pharmacol 2024; 181:4939-4957. [PMID: 38877789 DOI: 10.1111/bph.16470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/12/2024] [Accepted: 05/04/2024] [Indexed: 06/16/2024] Open
Abstract
Inflammatory signalling via the nod-like receptor (NLR) family pyrin domain-containing protein-3 (NLRP3) inflammasome has recently been implicated in the pathophysiology of atrial fibrillation (AF). However, the precise role of the NLRP3 inflammasome in various cardiac cell types is poorly understood. Targeting components or products of the inflammasome and preventing their proinflammatory consequences may constitute novel therapeutic treatment strategies for AF. In this review, we summarise the current understanding of the role of the inflammasome in AF pathogenesis. We first review the NLRP3 inflammasome pathway and inflammatory signalling in cardiomyocytes, (myo)fibroblasts and immune cells, such as neutrophils, macrophages and monocytes. Because numerous compounds targeting NLRP3 signalling are currently in preclinical development, or undergoing clinical evaluation for other indications than AF, we subsequently review known therapeutics, such as colchicine and canakinumab, targeting the NLRP3 inflammasome and evaluate their potential for treating AF.
Collapse
Affiliation(s)
- Alisha Niskala
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jordi Heijman
- Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
- Gottfried Schatz Research Center, Division of Medical Physics & Biophysics, Medical University of Graz, Graz, Austria
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Medicine and Research Center, Montréal Heart Institute and University de Montréal, Montréal, Canada
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arnela Saljic
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Zhang A, Wang X, Fan M, Guan Y, Jiang Y, Jin S, Liu L, Huangfu H, Song C. Lycopene alleviates zearalenone-induced oxidative stress, apoptosis, and NLRP3 inflammasome activation in mice kidneys. Toxicon 2024; 249:108078. [PMID: 39181415 DOI: 10.1016/j.toxicon.2024.108078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
The aim of this study was to investigate the protective effects of lycopene on renal damage caused by zearalenone (ZEN). Male Kunming mice were treated daily for 4 weeks by intragastric administration with 40 mg/kg ZEN in the presence or absence of lycopene (2.5 or 5 mg/kg). The results showed that lycopene markedly alleviated the damage of renal structure and function in mice induced by ZEN, as indicated by the reduced degree of pathological damage and the decreased levels of urea nitrogen and creatinine. Meanwhile, results of dihydroethidine (DHE) staining and biochemical markers revealed that ZEN exposure notably increased the levels of reactive oxygen species (ROS) and malondialdehyde (MDA), decreased the level of GSH, and reduced the activities of catalase (CAT) and superoxide dismutase (SOD). Administration of lycopene alleviated the increased oxidative stress induced by ZEN. Moreover, ZEN ingestion notably resulted in apoptosis, increased the protein levels of BCL2 associated X protein (Bax) and cleaved caspase-3, and decreased the protein levels of apoptosis regulator Bcl-2 (Bcl-2), which were reversed by lycopene intervention. Results of immunofluorescence demonstrated that lycopene reversed ZEN-induced the upregulation of NOD-like receptor pyrin domain-containing protein 3 (NLRP3), Caspase-1, and interleukin-1 beta (IL-1β) in mice kidneys. Lycopene supplementation could alleviate ZEN-induced renal toxicity by inhibiting oxidative stress, apoptosis, and NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Aiguo Zhang
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China
| | - Xuefei Wang
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China
| | - Mingming Fan
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China
| | - Yibo Guan
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China
| | - Yuchen Jiang
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China
| | - Shuangxing Jin
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China
| | - Lingling Liu
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China
| | - Heping Huangfu
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China
| | - Chao Song
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, No.6 Longzihu North road, Zhengzhou, Henan, 450046, China.
| |
Collapse
|
15
|
Ji L, Gao X, Xiao W, Yu S. Assessment of left atrial function provides incremental value: the left atrial volumetric/mechanical coupling index in patients with chronic kidney disease. Front Cardiovasc Med 2024; 11:1407531. [PMID: 39045007 PMCID: PMC11265283 DOI: 10.3389/fcvm.2024.1407531] [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: 03/26/2024] [Accepted: 06/14/2024] [Indexed: 07/25/2024] Open
Abstract
Background Heart failure is a common cause of adverse cardiovascular outcomes in patients with chronic kidney disease (CKD). Left atrial (LA) characteristics are thought to be involved in the development of heart failure. However, LA assessment is complex. Though a variety of parameters have been defined, there is no single parameter that best defines LA function. Pilot data indicate that left atrial volumetric/mechanical coupling index (LACI) may be useful, but data with CKD are lacking. Aim The objective of this study was to define LACI in a cohort of patients with CKD and to assess its value in evaluating LA function and predicting heart failure. Methods A cohort of patients with CKD was enrolled at our hospital between 2021 and 2023. Follow-up was performed for heart failure. LACI is a volumetric to mechanical coupling index, calculated as the ratio of the LA volume index to the tissue-Doppler myocardial velocity at atrial contraction. Spearman's rank correlation or Pearson's correlation was used to calculate the correlation between LACI and echocardiographic/hemodynamic variables. Receiver operating characteristic curve (ROC) analysis was utilised to derive the area under the curve (AUC) for LACI, LVGLS, LASr, LASct and LASI for the detection of heart failure. Kaplan-Meier survival curves were employed to compare clinical outcomes based on LACI thresholds. A multivariable logistic regression analysis was employed to assess the relationship between risk factors and elevated LACI. Cox proportional hazards regression was used to identify risk factors for heart failure. Results LACI showed a positive correlation with NT-proBNP, CK-MB, LAVI, E/e' and LASI (r = 0.504, 0.536, 0.856, 0.541 and 0.509, p < 0.001); and a negative correlation with LASr (r = -0.509, p < 0.001). On the ROC analysis for the determination of heart failure, the AUC of LACI was comparable to those of LVGLS (0.588 vs. 509, p = 0.464), LASr (0.588 vs. 0.448, p = 0.132), LASct (0.588 vs. 0.566, p = 0.971) and LASI (0.588 vs. 0.570, p = 0.874). The cardiovascular risk factors increased by LACI were age, BMI, diabetes, triglycerides, LA size, LASr, LASI, E/A, E/e' and EF (p < 0.05). During a median follow-up of 16 months (range, 6-28 months), the event-free survival curves demonstrated a higher risk of heart failure in the group with LACI > 5.0 (log-rank test: P < 0.001). LACI > 5.0 was an independent predictor of heart failure [OR: 0.121, 95% CI (0.020-0.740), p = 0.022]. Conclusion LACI may prove to be a valuable tool for assessing LA function in patients with CKD, and could be integrated into the routine assessment of LA for the purpose of prognostic assessment and clinical decision-making in patients with CKD.
Collapse
Affiliation(s)
- Liqin Ji
- Guizhou Medical University, Guiyang, Guizhou, China
| | - Xue Gao
- Guizhou Medical University, Guiyang, Guizhou, China
| | - Weiwei Xiao
- Guizhou Medical University, Guiyang, Guizhou, China
| | - Shaomei Yu
- Department of Ultrasound, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
16
|
Sahinoz M, Ikizler TA. Inflammasome activation: unraveling the link between chronic kidney disease and atrial fibrillation. Kidney Int 2024; 106:6-9. [PMID: 38906654 DOI: 10.1016/j.kint.2024.01.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/03/2024] [Indexed: 06/23/2024]
Affiliation(s)
- Melis Sahinoz
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - T Alp Ikizler
- Tennessee Valley Healthcare System, Nashville VA Medical Center, Nashville, Tennessee, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| |
Collapse
|
17
|
Lee KY, Lee SR, Choi EK, Choi J, Ahn HJ, Kwon S, Kim B, Han KD, Oh S, Lip GYH. Association between changes in renal function and clinical outcomes in anticoagulated atrial fibrillation patients with marginal renal function. A nationwide observational cohort study. Front Cardiovasc Med 2024; 11:1423336. [PMID: 38903967 PMCID: PMC11188773 DOI: 10.3389/fcvm.2024.1423336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 05/28/2024] [Indexed: 06/22/2024] Open
Abstract
Background Renal function is one of the crucial components for determining the dose and type of oral anticoagulants in atrial fibrillation (AF) patients, and is also closely associated with the risks of stroke and bleeding. This study aimed to assess renal function changes and their impact on clinical outcomes in anticoagulated AF patients with marginal renal function. Methods From a Korean claims database, patients with AF on anticoagulants and a baseline eGFR of 45 to <60 ml/min/1.73 m2 were studied. Patients were grouped by changes in renal function over two years-maintained, improved (eGFR >60 ml/min/1.73 m2), or worsened (eGFR <45 ml/min/1.73 m2)-the study analyzed outcomes including ischemic stroke, major bleeding, end-stage renal disease (ESRD), all-cause death, and a composite of clinical outcomes. Results A total of 5,126 patients were included in the study: 2,170 (42.3%) in the maintained group, 2,276 (44.4%) in the improved group, and 680 (13.1%) in the group with worsened renal function. The worsened group was older and had more prevalent comorbidities than other groups. After multivariable adjustment, the worsened group was associated with significantly higher risks of major bleeding (adjusted hazard ratio, 95% confidence interval; 1.46, 1.03-2.07, p = 0.035), ESRD (1.49, 1.24-1.80, p < 0.001), all-cause death (9.29, 4.92-17.6, p < 0.001), and the composite outcome (1.57, 1.36-1.83, p < 0.001). Conclusions In anticoagulated AF patients with marginal renal function, a substantial proportion of patients experienced renal function decline below eGFR 45 ml/min/1.73 m2 within 2 years. Renal function decline was associated with higher risks of major bleeding, ESRD, all-cause death, and the composite outcome compared to those who maintained their baseline renal function.
Collapse
Affiliation(s)
- Kyung-Yeon Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - So-Ryoung Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eue-Keun Choi
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - JungMin Choi
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyo-Jeong Ahn
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Soonil Kwon
- Division of Cardiology, Department of Internal Medicine, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Bongseong Kim
- Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Kyung-Do Han
- Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Seil Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Gregory Y. H. Lip
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Chest & Heart Hospital, Liverpool, United Kingdom
- Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
18
|
McCauley MD, Iacobellis G, Li N, Nattel S, Goldberger JJ. Targeting the Substrate for Atrial Fibrillation: JACC Review Topic of the Week. J Am Coll Cardiol 2024; 83:2015-2027. [PMID: 38749620 PMCID: PMC11460524 DOI: 10.1016/j.jacc.2024.02.050] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 06/17/2024]
Abstract
The identification of the pulmonary veins as a trigger source for atrial fibrillation (AF) has established pulmonary vein isolation (PVI) as a key target for AF ablation. However, PVI alone does not prevent recurrent AF in many patients, and numerous additional ablation strategies have failed to improve on PVI outcomes. This therapeutic limitation may be due, in part, to a failure to identify and intervene specifically on the pro-fibrillatory substrate within the atria and pulmonary veins. In this review paper, we highlight several emerging approaches with clinical potential that target atrial cardiomyopathy-the underlying anatomic, electrical, and/or autonomic disease affecting the atrium-in various stages of practice and investigation. In particular, we consider the evolving roles of risk factor modification, targeting of epicardial adipose tissue, tissue fibrosis, oxidative stress, and the inflammasome, along with aggressive early anti-AF therapy in AF management. Attention to combatting substrate development promises to improve outcomes in AF.
Collapse
Affiliation(s)
- Mark D McCauley
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA; Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA; Jesse Brown VA Medical Center, Chicago, Illinois, USA
| | - Gianluca Iacobellis
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Na Li
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, Texas, USA
| | - Stanley Nattel
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montreal, Quebec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany; IHU LIRYC and Fondation Bordeaux Université, Bordeaux, France
| | - Jeffrey J Goldberger
- Division of Cardiology, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
19
|
Zhao S, Hulsurkar MM, Lahiri SK, Aguilar-Sanchez Y, Munivez E, Müller FU, Jain A, Malovannaya A, Yiu CHK, Reilly S, Wehrens XHT. Atrial proteomic profiling reveals a switch towards profibrotic gene expression program in CREM-IbΔC-X mice with persistent atrial fibrillation. J Mol Cell Cardiol 2024; 190:1-12. [PMID: 38514002 DOI: 10.1016/j.yjmcc.2024.03.003] [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: 12/17/2023] [Revised: 02/19/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Overexpression of the CREM (cAMP response element-binding modulator) isoform CREM-IbΔC-X in transgenic mice (CREM-Tg) causes the age-dependent development of spontaneous AF. PURPOSE To identify key proteome signatures and biological processes accompanying the development of persistent AF through integrated proteomics and bioinformatics analysis. METHODS Atrial tissue samples from three CREM-Tg mice and three wild-type littermates were subjected to unbiased mass spectrometry-based quantitative proteomics, differential expression and pathway enrichment analysis, and protein-protein interaction (PPI) network analysis. RESULTS A total of 98 differentially expressed proteins were identified. Gene ontology analysis revealed enrichment for biological processes regulating actin cytoskeleton organization and extracellular matrix (ECM) dynamics. Changes in ITGAV, FBLN5, and LCP1 were identified as being relevant to atrial fibrosis and structural based on expression changes, co-expression patterns, and PPI network analysis. Comparative analysis with previously published datasets revealed a shift in protein expression patterns from ion-channel and metabolic regulators in young CREM-Tg mice to profibrotic remodeling factors in older CREM-Tg mice. Furthermore, older CREM-Tg mice exhibited protein expression patterns reminiscent of those seen in humans with persistent AF. CONCLUSIONS This study uncovered distinct temporal changes in atrial protein expression patterns with age in CREM-Tg mice consistent with the progressive evolution of AF. Future studies into the role of the key differentially abundant proteins identified in this study in AF progression may open new therapeutic avenues to control atrial fibrosis and substrate development in AF.
Collapse
Affiliation(s)
- Shuai Zhao
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mohit M Hulsurkar
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Satadru K Lahiri
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yuriana Aguilar-Sanchez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Elda Munivez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Frank Ulrich Müller
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Antrix Jain
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Anna Malovannaya
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA; Department of Biochemistry, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chi Him Kendrick Yiu
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, NIHR Oxford BRC, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, NIHR Oxford BRC, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine (in Cardiology), Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics (in Cardiology), Baylor College of Medicine, Houston, TX 77030, USA; Center for Space Medicine, Baylor College of Medicine, Houston, USA.
| |
Collapse
|
20
|
Tubeeckx MRL, De Keulenaer GW, Heidbuchel H, Segers VFM. Pathophysiology and clinical relevance of atrial myopathy. Basic Res Cardiol 2024; 119:215-242. [PMID: 38472506 DOI: 10.1007/s00395-024-01038-0] [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: 09/30/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 03/14/2024]
Abstract
Atrial myopathy is a condition that consists of electrical, structural, contractile, and autonomic remodeling of the atria and is the substrate for development of atrial fibrillation, the most common arrhythmia. Pathophysiologic mechanisms driving atrial myopathy are inflammation, oxidative stress, atrial stretch, and neurohormonal signals, e.g., angiotensin-II and aldosterone. These mechanisms initiate the structural and functional remodeling of the atrial myocardium. Novel therapeutic strategies are being developed that target the pathophysiologic mechanisms of atrial myopathy. In this review, we will discuss the pathophysiology of atrial myopathy, as well as diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Michiel R L Tubeeckx
- Laboratory of Physiopharmacology, Universiteitsplein 1, Building T (2nd Floor), 2610, Antwerp, Belgium.
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, Universiteitsplein 1, Building T (2nd Floor), 2610, Antwerp, Belgium
- Department of Cardiology, ZNA Middelheim Hospital Antwerp, Antwerp, Belgium
| | - Hein Heidbuchel
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium
| | - Vincent F M Segers
- Laboratory of Physiopharmacology, Universiteitsplein 1, Building T (2nd Floor), 2610, Antwerp, Belgium
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium
| |
Collapse
|
21
|
Huang M, Huiskes FG, de Groot NMS, Brundel BJJM. The Role of Immune Cells Driving Electropathology and Atrial Fibrillation. Cells 2024; 13:311. [PMID: 38391924 PMCID: PMC10886649 DOI: 10.3390/cells13040311] [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: 12/20/2023] [Revised: 02/02/2024] [Accepted: 02/04/2024] [Indexed: 02/24/2024] Open
Abstract
Atrial fibrillation (AF) is the most common progressive cardiac arrhythmia worldwide and entails serious complications including stroke and heart failure. Despite decades of clinical research, the current treatment of AF is suboptimal. This is due to a lack of knowledge on the mechanistic root causes of AF. Prevailing theories indicate a key role for molecular and structural changes in driving electrical conduction abnormalities in the atria and as such triggering AF. Emerging evidence indicates the role of the altered atrial and systemic immune landscape in driving this so-called electropathology. Immune cells and immune markers play a central role in immune remodeling by exhibiting dual facets. While the activation and recruitment of immune cells contribute to maintaining atrial stability, the excessive activation and pronounced expression of immune markers can foster AF. This review delineates shifts in cardiac composition and the distribution of immune cells in the context of cardiac health and disease, especially AF. A comprehensive exploration of the functions of diverse immune cell types in AF and other cardiac diseases is essential to unravel the intricacies of immune remodeling. Usltimately, we delve into clinical evidence showcasing immune modifications in both the atrial and systemic domains among AF patients, aiming to elucidate immune markers for therapy and diagnostics.
Collapse
Affiliation(s)
- Mingxin Huang
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, The Netherlands; (M.H.); (F.G.H.)
- Department of Cardiology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Fabries G. Huiskes
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, The Netherlands; (M.H.); (F.G.H.)
| | | | - Bianca J. J. M. Brundel
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, The Netherlands; (M.H.); (F.G.H.)
| |
Collapse
|
22
|
Zhao S, Hulsurkar MM, Lahiri SK, Aguilar-Sanchez Y, Munivez E, Müller FU, Jain A, Malovannaya A, Yiu K, Reilly S, Wehrens XH. Atrial Proteomic Profiling Reveals a Switch Towards Profibrotic Gene Expression Program in CREM-IbΔC-X Mice with Persistent Atrial Fibrillation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.575097. [PMID: 38260363 PMCID: PMC10802622 DOI: 10.1101/2024.01.10.575097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background Overexpression of the CREM (cAMP response element-binding modulator) isoform CREM-IbΔC-X in transgenic mice (CREM-Tg) causes the age-dependent development of spontaneous AF. Purpose To identify key proteome signatures and biological processes accompanying the development of persistent AF through integrated proteomics and bioinformatics analysis. Methods Atrial tissue samples from three CREM-Tg mice and three wild-type littermates were subjected to unbiased mass spectrometry-based quantitative proteomics, differential expression and pathway enrichment analysis, and protein-protein interaction (PPI) network analysis. Results A total of 98 differentially expressed proteins were identified. Gene ontology analysis revealed enrichment for biological processes regulating actin cytoskeleton organization and extracellular matrix (ECM) dynamics. Changes in ITGAV, FBLN5, and LCP1 were identified as being relevant to atrial fibrosis and remodeling based on expression changes, co-expression patterns, and PPI network analysis. Comparative analysis with previously published datasets revealed a shift in protein expression patterns from ion-channel and metabolic regulators in young CREM-Tg mice to profibrotic remodeling factors in older CREM-Tg mice. Furthermore, older CREM-Tg mice exhibited protein expression patterns that resembled those of humans with persistent AF. Conclusions This study uncovered distinct temporal changes in atrial protein expression patterns with age in CREM-Tg mice consistent with the progressive evolution of AF. Future studies into the role of the key differentially abundant proteins identified in this study in AF progression may open new therapeutic avenues to control atrial fibrosis and substrate development in AF.
Collapse
Affiliation(s)
- Shuai Zhao
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mohit M. Hulsurkar
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Satadru K. Lahiri
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yuriana Aguilar-Sanchez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Elda Munivez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Frank Ulrich Müller
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Antrix Jain
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Anna Malovannaya
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
- Department of Biochemistry, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kendrick Yiu
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, NIHR Oxford BRC, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, NIHR Oxford BRC, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Xander H.T. Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine (in Cardiology), Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics (in Cardiology), Baylor College of Medicine, Houston, TX 77030, USA
- Center for Space Medicine, Baylor College of Medicine, Houston, USA
| |
Collapse
|
23
|
Song J, Wu J, Robichaux DJ, Li T, Wang S, Arredondo Sancristobal MJ, Dong B, Dobrev D, Karch J, Thomas SS, Li N. A High-Protein Diet Promotes Atrial Arrhythmogenesis via Absent-in-Melanoma 2 Inflammasome. Cells 2024; 13:108. [PMID: 38247800 PMCID: PMC10814244 DOI: 10.3390/cells13020108] [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: 12/17/2023] [Revised: 12/29/2023] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
High-protein diets (HPDs) offer health benefits, such as weight management and improved metabolic profiles. The effects of HPD on cardiac arrhythmogenesis remain unclear. Atrial fibrillation (AF), the most common arrhythmia, is associated with inflammasome activation. The role of the Absent-in-Melanoma 2 (AIM2) inflammasome in AF pathogenesis remains unexplored. In this study, we discovered that HPD increased susceptibility to AF. To demonstrate the involvement of AIM2 signaling in the pathogenesis of HPD-induced AF, wildtype (WT) and Aim2-/- mice were fed normal-chow (NC) and HPD, respectively. Four weeks later, inflammasome activity was upregulated in the atria of WT-HPD mice, but not in the Aim2-/--HPD mice. The increased AF vulnerability in WT-HPD mice was associated with abnormal sarcoplasmic reticulum (SR) Ca2+-release events in atrial myocytes. HPD increased the cytoplasmic double-strand (ds) DNA level, causing AIM2 activation. Genetic inhibition of AIM2 in Aim2-/- mice reduced susceptibility to AF, cytoplasmic dsDNA level, mitochondrial ROS production, and abnormal SR Ca2+-release in atrial myocytes. These data suggest that HPD creates a substrate conducive to AF development by activating the AIM2-inflammasome, which is associated with mitochondrial oxidative stress along with proarrhythmic SR Ca2+-release. Our data imply that targeting the AIM2 inflammasome might constitute a novel anti-AF strategy in certain patient subpopulations.
Collapse
Affiliation(s)
- Jia Song
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX 77030, USA (M.J.A.S.)
| | - Jiao Wu
- Department of Medicine, Section of Nephrology, Houston, TX 77030, USA
| | - Dexter J. Robichaux
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA (D.D.)
| | - Tingting Li
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX 77030, USA (M.J.A.S.)
| | - Shuyue Wang
- Department of Medicine, Section of Gastroenterology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Bingning Dong
- Department of Medicine, Section of Gastroenterology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dobromir Dobrev
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA (D.D.)
- Institute of Pharmacology, University Duisburg-Essen, 45147 Essen, Germany
- Department of Medicine, Montreal Heart Institute, Université de Montréal, Montréal, QC H1T 1C8, Canada
| | - Jason Karch
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA (D.D.)
| | - Sandhya S. Thomas
- Department of Medicine, Section of Nephrology, Houston, TX 77030, USA
- Michael E. Debakey VA Medical Center, Houston, TX 77030, USA
| | - Na Li
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX 77030, USA (M.J.A.S.)
| |
Collapse
|
24
|
Chen Z, Cheng Z, Ding C, Cao T, Chen L, Wang H, Li J, Huang X. ROS-Activated TRPM2 Channel: Calcium Homeostasis in Cardiovascular/renal System and Speculation in Cardiorenal Syndrome. Cardiovasc Drugs Ther 2023:10.1007/s10557-023-07531-3. [PMID: 38108918 DOI: 10.1007/s10557-023-07531-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 12/19/2023]
Abstract
The transient receptor potential melastatin 2 (TRPM2) channel is a nonselective calcium channel that is sensitive to oxidative stress (OS), and is widely expressed in multiple organs, such as the heart, kidney, and brain, which is inextricably related to calcium dyshomeostasis and downstream pathological events. Due to the increasing global burden of kidney or cardiovascular diseases (CVDs), safe and efficient drugs specific to novel targets are imperatively needed. Notably, investigation of the possibility to regard the TRPM2 channel as a new therapeutic target in ROS-related CVDs or renal diseases is urgently required because the roles of the TRPM2 channel in heart or kidney diseases have not received enough attention and thus have not been fully elaborated. Therefore, we aimed to review the involvement of the TRPM2 channel in cardiovascular disorders related to kidney or typical renal diseases and attempted to speculate about TRPM2-mediated mechanisms of cardiorenal syndrome (CRS) to provide representative perspectives for future research about novel and effective therapeutic strategies.
Collapse
Affiliation(s)
- Zihan Chen
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Queen Mary School, Medical Department, Nanchang University, Nanchang, China
| | - Zaihua Cheng
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Congcong Ding
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tianyu Cao
- Biological anthropology, University of California, Santa Barbara, CA, USA
| | - Ling Chen
- Department of Cardiology, the First People's Hospital of Jiujiang, Jiujiang, China
| | - Hong Wang
- Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Junpei Li
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Xiao Huang
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|