1
|
Drăgoi CM, Diaconu CC, Nicolae AC, Dumitrescu IB. Redox Homeostasis and Molecular Biomarkers in Precision Therapy for Cardiovascular Diseases. Antioxidants (Basel) 2024; 13:1163. [PMID: 39456418 PMCID: PMC11504313 DOI: 10.3390/antiox13101163] [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: 07/31/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Precision medicine is envisioned as the future of cardiovascular healthcare, offering a more tailored and effective method for managing cardiovascular diseases compared to the traditional one-size-fits-all approaches. The complex role of oxidative stress in chronic diseases within the framework of precision medicine was carefully explored, delving into the cellular redox status and its critical involvement in the pathophysiological complexity of cardiovascular diseases (CVDs). The review outlines the mechanisms of reactive oxygen species generation and the function of antioxidants in maintaining redox balance. It emphasizes the elevated reactive oxygen species concentrations observed in heart failure and their detrimental impact on cardiovascular health. Various sources of ROS within the cardiovascular system are examined, including mitochondrial dysfunction, which contributes to oxidative stress and mitochondrial DNA degradation. The article also addresses oxidative stress's role in myocardial remodeling, a process pivotal to the progression of heart diseases. By integrating these aspects, the review underscores the importance of redox homeostasis and identifies molecular biomarkers that can enhance precision therapy for CVDs. The insights provided aim to pave the way for targeted therapeutic strategies that mitigate oxidative stress, thereby improving patient outcomes in cardiovascular medicine.
Collapse
Affiliation(s)
- Cristina Manuela Drăgoi
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania; (C.M.D.); (I.-B.D.)
| | - Camelia Cristina Diaconu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania;
- Department of Internal Medicine, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| | - Alina Crenguța Nicolae
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania; (C.M.D.); (I.-B.D.)
| | - Ion-Bogdan Dumitrescu
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania; (C.M.D.); (I.-B.D.)
| |
Collapse
|
2
|
Mauriello A, Ascrizzi A, Roma AS, Molinari R, Caturano A, Imbalzano E, D’Andrea A, Russo V. Effects of Heart Failure Therapies on Atrial Fibrillation: Biological and Clinical Perspectives. Antioxidants (Basel) 2024; 13:806. [PMID: 39061875 PMCID: PMC11273474 DOI: 10.3390/antiox13070806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/21/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Heart failure (HF) and atrial fibrillation (AF) are prevalent cardiovascular diseases that contribute significantly to morbidity, mortality, hospitalisation, and healthcare costs. It is not uncommon for these conditions to coexist and have mutually reinforcing effects. A critical factor in the aetiology of these conditions is oxidative stress, driven by reactive oxygen species (ROS), which contributes to atrial remodelling and fibrosis. The recent introduction of new drugs for the treatment of heart failure has also had an impact on the management of atrial fibrillation due to their influence on oxidative stress. The objective of this review is to analyse the effects of these therapies, including their role in mitigating ROS, on the prevention and treatment of AF in HF patients.
Collapse
Affiliation(s)
- Alfredo Mauriello
- Cardiology Unit, Department of Medical and Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80131 Naples, Italy; (A.M.); (A.A.); (A.S.R.); (R.M.)
- Cardiology and Intensive Care Unit, Department of Cardiology, Umberto I Hospital, 84014 Nocera Inferiore, Italy;
| | - Antonia Ascrizzi
- Cardiology Unit, Department of Medical and Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80131 Naples, Italy; (A.M.); (A.A.); (A.S.R.); (R.M.)
| | - Anna Selvaggia Roma
- Cardiology Unit, Department of Medical and Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80131 Naples, Italy; (A.M.); (A.A.); (A.S.R.); (R.M.)
| | - Riccardo Molinari
- Cardiology Unit, Department of Medical and Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80131 Naples, Italy; (A.M.); (A.A.); (A.S.R.); (R.M.)
| | - Alfredo Caturano
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy;
| | - Egidio Imbalzano
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy;
| | - Antonello D’Andrea
- Cardiology and Intensive Care Unit, Department of Cardiology, Umberto I Hospital, 84014 Nocera Inferiore, Italy;
| | - Vincenzo Russo
- Cardiology Unit, Department of Medical and Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80131 Naples, Italy; (A.M.); (A.A.); (A.S.R.); (R.M.)
| |
Collapse
|
3
|
Zhang J, Han Y, Jia R, Zhu Q, Wang X, Liu M, Zhang W. Exploring the role of myeloperoxidase in the atherosclerotic process in hypoxic mice based on the MAPK signaling pathway. Biochem Pharmacol 2024; 225:116275. [PMID: 38729447 DOI: 10.1016/j.bcp.2024.116275] [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: 12/24/2023] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
Atherosclerosis (AS) is the common pathophysiological basis of various cardiovascular diseases and the leading cause of death from cardiovascular disease worldwide. When the body is in a hypoxic environment, enhanced oxidative stress and significant accumulation of reactive oxygen species (ROS) in tissue cells exacerbate the inflammatory response, resulting in increased release of myeloperoxidase (MPO), catalyzing the formation of large quantities of hypochlorous acid (HOCl), further oxidative modification of low-density lipoprotein (LDL), and exacerbating the formation and progression of atherosclerotic plaques. The MAPK signaling pathway is important in oxidative stress-mediated promotion of atherogenesis. MPO -/- mice were used in this study to establish a hypoxia model simulating 5000 m altitude and a Western high-fat diet-induced atherosclerosis model for 12 weeks. Exploring the role of MPO in the atherosclerotic process in hypoxic mice by observing the MAPK signaling pathway to provide a therapeutic target for the prevention and treatment of hypoxic atherosclerotic disease in the plateau. We found that hypoxia promotes the formation of atherosclerosis in mice, and the mechanism may be that increased MPO in vivo promotes an inflammatory response, which plays a crucial role in the formation of atherosclerosis. In addition, hypoxia further exacerbates plaque instability by activating the MAPK signaling pathway to upregulate vascular endothelial growth factor (VEGF) and matrix metalloproteinase-9 (MMP9), which in turn promotes angiogenesis within the plaque. Therefore, a potential target for preventing and treating hypoxic atherosclerotic disease is the inhibition of MPO.
Collapse
Affiliation(s)
- Jingxuan Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Ying Han
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Ruhan Jia
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Qinfang Zhu
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China; Qinghai Provincial People's Hospital, Xining, Qinghai, China
| | - Xiaozhou Wang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China; Department of Hypertension, Qinghai Cardio-Cerebrovascular Hospital, Xining, Qinghai, China
| | - Meiheng Liu
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Wei Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China.
| |
Collapse
|
4
|
Ma XG, Chen R. Letter: Systemic Immune-Inflammatory Index as a Novel Biomarker for Predicting the Development of Arrhythmia. Angiology 2024:33197241256994. [PMID: 38789281 DOI: 10.1177/00033197241256994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Affiliation(s)
- Xin-Guo Ma
- Department of ECG, Liaocheng People's Hospital Affiliated to Shandong First Medical University, Liaocheng, PR China
| | - Ran Chen
- Department of ECG, Liaocheng People's Hospital Affiliated to Shandong First Medical University, Liaocheng, PR China
| |
Collapse
|
5
|
Lei M, Salvage SC, Jackson AP, Huang CLH. Cardiac arrhythmogenesis: roles of ion channels and their functional modification. Front Physiol 2024; 15:1342761. [PMID: 38505707 PMCID: PMC10949183 DOI: 10.3389/fphys.2024.1342761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/22/2024] [Indexed: 03/21/2024] Open
Abstract
Cardiac arrhythmias cause significant morbidity and mortality and pose a major public health problem. They arise from disruptions in the normally orderly propagation of cardiac electrophysiological activation and recovery through successive cardiomyocytes in the heart. They reflect abnormalities in automaticity, initiation, conduction, or recovery in cardiomyocyte excitation. The latter properties are dependent on surface membrane electrophysiological mechanisms underlying the cardiac action potential. Their disruption results from spatial or temporal instabilities and heterogeneities in the generation and propagation of cellular excitation. These arise from abnormal function in their underlying surface membrane, ion channels, and transporters, as well as the interactions between them. The latter, in turn, form common regulatory targets for the hierarchical network of diverse signaling mechanisms reviewed here. In addition to direct molecular-level pharmacological or physiological actions on these surface membrane biomolecules, accessory, adhesion, signal transduction, and cytoskeletal anchoring proteins modify both their properties and localization. At the cellular level of excitation-contraction coupling processes, Ca2+ homeostatic and phosphorylation processes affect channel activity and membrane excitability directly or through intermediate signaling. Systems-level autonomic cellular signaling exerts both acute channel and longer-term actions on channel expression. Further upstream intermediaries from metabolic changes modulate the channels both themselves and through modifying Ca2+ homeostasis. Finally, longer-term organ-level inflammatory and structural changes, such as fibrotic and hypertrophic remodeling, similarly can influence all these physiological processes with potential pro-arrhythmic consequences. These normal physiological processes may target either individual or groups of ionic channel species and alter with particular pathological conditions. They are also potentially alterable by direct pharmacological action, or effects on longer-term targets modifying protein or cofactor structure, expression, or localization. Their participating specific biomolecules, often clarified in experimental genetically modified models, thus constitute potential therapeutic targets. The insights clarified by the physiological and pharmacological framework outlined here provide a basis for a recent modernized drug classification. Together, they offer a translational framework for current drug understanding. This would facilitate future mechanistically directed therapeutic advances, for which a number of examples are considered here. The latter are potentially useful for treating cardiac, in particular arrhythmic, disease.
Collapse
Affiliation(s)
- Ming Lei
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Samantha C. Salvage
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Antony P. Jackson
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Christopher L.-H. Huang
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
6
|
Xu Z, Rao X, Xing Y, Zhu Z, Yan L, Huang J, Zhang J, Zheng R. Connecting atrial fibrillation to digestive neoplasms: exploring mediation via ischemic stroke and heart failure in Mendelian randomization studies. Front Oncol 2024; 14:1301327. [PMID: 38444673 PMCID: PMC10912520 DOI: 10.3389/fonc.2024.1301327] [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: 09/24/2023] [Accepted: 01/22/2024] [Indexed: 03/07/2024] Open
Abstract
Background Notwithstanding the acknowledged interplay between atrial fibrillation (AF) and the emergence of digestive system neoplasms, the intricacies of this relationship remain ambiguous. By capitalizing univariable Mendelian Randomization (MR) complemented by a mediated MR tactic, our pursuit was to elucidate the causative roles of AF in precipitating digestive system malignancies and potential intermediary pathways. Method This research endeavor seeks to scrutinize the causal clinical implications of whether genetic predispositions to AF correlate with an increased risk of digestive system malignancies, employing MR analytical techniques. Utilizing a dataset amalgamated from six studies related to AF, encompassing over 1,000,000 subjects, we performed univariable MR assessments, employing the random-effects inverse-variance weighted (IVW) methodology as our principal analytical paradigm. Subsequently, a mediated MR framework was employed to probe the potential mediating influence of AF on the nexus between hypertension (HT), heart failure (HF), ischemic stroke (IS), coronary artery disease (CAD), and digestive system neoplasms. Result The univariable MR evaluation unveiled a notable causal nexus between the genetic inclination toward AF and the genetic susceptibility to colon, esophageal, and small intestine malignancies. The mediated MR scrutiny ascertained that the genetic inclination for AF amplifies the risk profile for colon cancer via IS pathways and partially explains the susceptibility to esophageal and small intestine tumors through the HF pathway. Conclusion Our investigative endeavor has highlighted a definitive causative association between genetic inclination to AF and specific digestive system neoplasms, spotlighting IS and HF as instrumental mediators. Such revelations furnish pivotal perspectives on the complex genetic interconnections between cardiovascular anomalies and certain digestive tract tumors, emphasizing prospective therapeutic and diagnostic worthy of pursuit.
Collapse
Affiliation(s)
- Zhijie Xu
- Beijing University of Chinese Medicine, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xuezhi Rao
- Beijing University of Chinese Medicine, Beijing, China
- The Second School of Clinical Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yaxuan Xing
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhiwei Zhu
- Beijing University of Chinese Medicine, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Longmei Yan
- Beijing University of Chinese Medicine, Beijing, China
| | - Jian Huang
- Department of Acupuncture and Moxibustion, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jingchun Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruwen Zheng
- Department of Acupuncture and Moxibustion, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
7
|
Chui PW, Khokhar A, Gordon KS, Dziura J, Burg MM, Brandt C, Haskell SG, Malm B, Bastian LA, Gandhi PU. Opioid prescription and risk of atrial fibrillation in younger veterans. Am Heart J 2024; 268:61-67. [PMID: 37949420 DOI: 10.1016/j.ahj.2023.11.001] [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: 08/07/2023] [Revised: 10/30/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Opioids may play a part in the development of atrial fibrillation (AF). Understanding the relationship between opioid exposure and AF can help providers better assess the risk and benefits of prescribing opioids. OBJECTIVE To assess the incidence of AF as a function of prescribed opioids and opioid type. DESIGN We performed unadjusted and adjusted time-updated Cox regressions to assess the association between opioid exposure and incident AF. PARTICIPANTS The national study sample was comprised of Veterans enrolled in the Veterans Health Administration (VHA) who served in support of post-9/11 operations. MAIN MEASURES The main predictor of interest was prescription opioid exposure, which was treated as a time-dependent variable. The first was any opioid exposure (yes/no). Secondary was opioid type. The outcome, incident AF, was identified through ICD-9-CM diagnostic codes at any primary care visit after the baseline period. KEY RESULTS A total of 609,763 veterans (mean age: 34 years and 13.24% female) were included in our study. Median follow-up time was 4.8 years. Within this cohort, 124,395 veterans (20.40%) were prescribed an opioid. A total of 1,455 Veterans (0.24%) were diagnosed with AF. In adjusted time-updated Cox regressions, the risk of incident AF was higher in the veterans prescribed opioids (hazard ratio [HR]: 1.47; 95% confidence interval [CI]: 1.38-1.57). In adjusted time-updated Cox regressions, both immunomodulating and nonimmunomodulating opioid type was associated with increased risk of incident AF (HR: 1.40; 95% CI: 1.25-1.57 and HR: 1.49; 95% CI: 1.39-1.60), compared to no opioid use, respectively. CONCLUSIONS Our findings suggest opioid prescription may be a modifiable risk factor for the development of AF.
Collapse
Affiliation(s)
- Philip W Chui
- Kaiser Los Angeles Medical Center, Section of Cardiology, Los Angeles, CA; Southern California Heart Center, San Gabriel, CA
| | - Arshjot Khokhar
- Section of General Internal Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Kirsha S Gordon
- Pain Research, Informatics, Multimorbidities, and Education (PRIME) Center, VA Connecticut Healthcare System, West Haven, CT; Yale Center for Analytical Sciences, School of Public Health, Yale University School of Medicine, New Haven, CT
| | - James Dziura
- Yale Center for Analytical Sciences, School of Public Health, Yale University School of Medicine, New Haven, CT
| | - Matthew M Burg
- VA Connecticut Health System, West Haven, CT; Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Cynthia Brandt
- Pain Research, Informatics, Multimorbidities, and Education (PRIME) Center, VA Connecticut Healthcare System, West Haven, CT
| | - Sally G Haskell
- Pain Research, Informatics, Multimorbidities, and Education (PRIME) Center, VA Connecticut Healthcare System, West Haven, CT; Section of General Internal Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Brian Malm
- VA Connecticut Health System, West Haven, CT; Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Lori A Bastian
- Pain Research, Informatics, Multimorbidities, and Education (PRIME) Center, VA Connecticut Healthcare System, West Haven, CT; Section of General Internal Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT.
| | - Parul U Gandhi
- VA Connecticut Health System, West Haven, CT; Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
8
|
Binek A, Castans C, Jorge I, Bagwan N, Rodríguez JM, Fernández-Jiménez R, Galán-Arriola C, Oliver E, Gómez M, Clemente-Moragón A, Ibanez B, Camafeita E, Vázquez J. Oxidative Post-translational Protein Modifications upon Ischemia/Reperfusion Injury. Antioxidants (Basel) 2024; 13:106. [PMID: 38247530 PMCID: PMC10812827 DOI: 10.3390/antiox13010106] [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: 11/29/2023] [Revised: 12/30/2023] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
While reperfusion, or restoration of coronary blood flow in acute myocardial infarction, is a requisite for myocardial salvage, it can paradoxically induce a specific damage known as ischemia/reperfusion (I/R) injury. Our understanding of the precise pathophysiological molecular alterations leading to I/R remains limited. In this study, we conducted a comprehensive and unbiased time-course analysis of post-translational modifications (PTMs) in the post-reperfused myocardium of two different animal models (pig and mouse) and evaluated the effect of two different cardioprotective therapies (ischemic preconditioning and neutrophil depletion). In pigs, a first wave of irreversible oxidative damage was observed at the earliest reperfusion time (20 min), impacting proteins essential for cardiac contraction. A second wave, characterized by irreversible oxidation on different residues and reversible Cys oxidation, occurred at late stages (6-12 h), affecting mitochondrial, sarcomere, and inflammation-related proteins. Ischemic preconditioning mitigated the I/R damage caused by the late oxidative wave. In the mouse model, the two-phase pattern of oxidative damage was replicated, and neutrophil depletion mitigated the late wave of I/R-related damage by preventing both Cys reversible oxidation and irreversible oxidation. Altogether, these data identify protein PTMs occurring late after reperfusion as an actionable therapeutic target to reduce the impact of I/R injury.
Collapse
Grants
- PGC2018-097019-B-I00, PID2021-122348NB-I00, PID2022-140176OB-I00 Spanish Ministry of Science, Innovation and Universities
- Fondo de Investigación Sanitaria grant PRB3 PT17/0019/0003- ISCIII-SGEFI / ERDF, ProteoRed Instituto de Salud Carlos III
- IMMUNO-VAR, P2022/BMD-7333, and RENIM-CM, P2022/BMD-7403 Comunidad de Madrid
- HR17-00247, HR22-00533 and HR22-00253 "la Caixa" Banking Foundation
- ERC Consolidator Grant "MATRIX", 819775 European Commission
- grant PI22/01560 ISCIII-Fondo de Investigación Sanitaria and European Union
- FP7-PEOPLE-2013-ITN-Cardionext European Union's Seventh Framework Programme
- Formacion del Profesorado Universitario (FPU14/05292) Spanish Ministry of Education, Culture and Sports
- PID2021-133167OB-100, RYC2020-028884-I, CEX2020-001041-S MCIN/AEI/10.13039/501100011033
Collapse
Affiliation(s)
- Aleksandra Binek
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Celia Castans
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Inmaculada Jorge
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Navratan Bagwan
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - José Manuel Rodríguez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Rodrigo Fernández-Jiménez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Department of Cardiology, Hospital Universitario Clínico San Carlos, Profesor Martín Lagos, s/n, 28040 Madrid, Spain
| | - Carlos Galán-Arriola
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Eduardo Oliver
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Mónica Gómez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Agustín Clemente-Moragón
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- IIS-Fundación Jiménez Díaz Hospital, Avenida Reyes Católicos, 2, 28040 Madrid, Spain
| | - Emilio Camafeita
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| |
Collapse
|
9
|
Tong F, Sun Z. Identification and validation of potential biomarkers for atrial fibrillation based on integrated bioinformatics analysis. Front Cell Dev Biol 2024; 11:1190273. [PMID: 38274270 PMCID: PMC10808641 DOI: 10.3389/fcell.2023.1190273] [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: 03/20/2023] [Accepted: 12/29/2023] [Indexed: 01/27/2024] Open
Abstract
Background: Globally, the most common form of arrhythmias is atrial fibrillation (AF), which causes severe morbidity, mortality, and socioeconomic burden. The application of machine learning algorithms in combination with weighted gene co-expression network analysis (WGCNA) can be used to screen genes, therefore, we aimed to screen for potential biomarkers associated with AF development using this integrated bioinformatics approach. Methods: On the basis of the AF endocardium gene expression profiles GSE79768 and GSE115574 from the Gene Expression Omnibus database, differentially expressed genes (DEGs) between AF and sinus rhythm samples were identified. DEGs enrichment analysis and transcription factor screening were then performed. Hub genes for AF were screened using WGCNA and machine learning algorithms, and the diagnostic accuracy was assessed by the receiver operating characteristic (ROC) curves. GSE41177 was used as the validation set for verification. Subsequently, we identified the specific signaling pathways in which the key biomarkers were involved, using gene set enrichment analysis and reverse prediction of mRNA-miRNA interaction pairs. Finally, we explored the associations between the hub genes and immune microenvironment and immune regulation. Results: Fifty-seven DEGs were identified, and the two hub genes, hypoxia inducible factor 1 subunit alpha inhibitor (HIF1AN) and mitochondrial inner membrane protein MPV17 (MPV17), were screened using WGCNA combined with machine learning algorithms. The areas under the receiver operating characteristic curves for MPV17 and HIF1AN validated that two genes predicted AF development, and the differential expression of the hub genes was verified in the external validation dataset. Enrichment analysis showed that MPV17 and HIF1AN affect mitochondrial dysfunction, oxidative stress, gap junctions, and other signaling pathway functions. Immune cell infiltration and immunomodulatory correlation analyses showed that MPV17 and HIF1AN are strongly correlated with the content of immune cells and significantly correlated with HLA expression. Conclusion: The identification of hub genes associated with AF using WGCNA combined with machine learning algorithms and their correlation with immune cells and immune gene expression can elucidate the molecular mechanisms underlying AF occurrence. This may further identify more accurate and effective biomarkers and therapeutic targets for the diagnosis and treatment of AF.
Collapse
Affiliation(s)
| | - Zhijun Sun
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
10
|
Liu J, Lin C, Zhou T, Bao Y, Xie Y, Wei Y, Luo Q, Ling T, Pan W, Zhang N, Lu L, Wu L, Jin Q. Plasma myeloperoxidase: association with atrial fibrillation progression and recurrence after catheter ablation. Front Cardiovasc Med 2023; 10:1150324. [PMID: 37719981 PMCID: PMC10501448 DOI: 10.3389/fcvm.2023.1150324] [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: 01/24/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023] Open
Abstract
Background Myeloperoxidase (MPO), released by activated neutrophils, is significantly increased in atrial fibrillation (AF). MPO may play a role in the progression of atrial fibrillation and further involved in AF recurrence after catheter ablation. We compared plasma MPO levels in paroxysmal and persistent AF and explored their role in AF recurrence after catheter ablation. Methods Plasma MPO levels were measured in consecutive patients with paroxysmal AF (n = 225) and persistent AF (n = 106). Samples of patients were collected from the femoral vein during catheter ablation and all patients included were followed up after catheter ablation. Results Plasma MPO levels increased from paroxysmal AF to persistent AF patients (56.31 [40.33-73.51] vs. 64.11 [48.65-81.11] ng/ml, p < 0.001). MPO significantly correlated with left atrium volume (LAV) and there existed a significant interaction between the two in relation to AF recurrence (p for interaction <0.05). During a median follow-up of 14 months, 28 patients with paroxysmal AF (12.44%) and 27 patients with persistent AF (25.47%) presented with recurrence after catheter ablation. The percentage of recurrence increased stepwise with increasing tertiles of MPO levels in both paroxysmal AF and persistent AF. MPO levels remained independently associated with AF recurrence after adjusting for potential confounding variables. Conclusion MPO levels were higher in persistent AF than in paroxysmal AF and MPO was positively correlated with LAV in AF. Elevated MPO levels may predispose a switch in AF phenotype and AF recurrence after catheter ablation.
Collapse
Affiliation(s)
- Jingmeng Liu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changjian Lin
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Taojie Zhou
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yangyang Bao
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun Xie
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Wei
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingzhi Luo
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyou Ling
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenqi Pan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Lu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liqun Wu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Jin
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Wang Y, Wang Q, Liu P, Jin L, Qin X, Zheng Q. Construction and validation of a cuproptosis-related diagnostic gene signature for atrial fibrillation based on ensemble learning. Hereditas 2023; 160:34. [PMID: 37620966 PMCID: PMC10464108 DOI: 10.1186/s41065-023-00297-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/02/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common type of cardiac arrhythmia. Nonetheless, the accurate diagnosis of this condition continues to pose a challenge when relying on conventional diagnostic techniques. Cell death is a key factor in the pathogenesis of AF. Existing investigations suggest that cuproptosis may also contribute to AF. This investigation aimed to identify a novel diagnostic gene signature associated with cuproptosis for AF using ensemble learning methods and discover the connection between AF and cuproptosis. RESULTS Two genes connected to cuproptosis, including solute carrier family 31 member 1 (SLC31A1) and lipoic acid synthetase (LIAS), were selected by integration of random forests and eXtreme Gradient Boosting algorithms. Subsequently, a diagnostic model was constructed that includes the two genes for AF using the Light Gradient Boosting Machine (LightGBM) algorithm with good performance (the area under the curve value > 0.75). The microRNA-transcription factor-messenger RNA network revealed that homeobox A9 (HOXA9) and Tet methylcytosine dioxygenase 1 (TET1) could target SLC31A1 and LIAS in AF. Functional enrichment analysis indicated that cuproptosis might be connected to immunocyte activities. Immunocyte infiltration analysis using the CIBERSORT algorithm suggested a greater level of neutrophils in the AF group. According to the outcomes of Spearman's rank correlation analysis, there was a negative relation between SLC31A1 and resting dendritic cells and eosinophils. The study found a positive relationship between LIAS and eosinophils along with resting memory CD4+ T cells. Conversely, a negative correlation was detected between LIAS and CD8+ T cells and regulatory T cells. CONCLUSIONS This study successfully constructed a cuproptosis-related diagnostic model for AF based on the LightGBM algorithm and validated its diagnostic efficacy. Cuproptosis may be regulated by HOXA9 and TET1 in AF. Cuproptosis might interact with infiltrating immunocytes in AF.
Collapse
Affiliation(s)
- Yixin Wang
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiaozhu Wang
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Peng Liu
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lingyan Jin
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinghua Qin
- Xi'an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.
| | - Qiangsun Zheng
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
12
|
Huang CLH, Lei M. Cardiomyocyte electrophysiology and its modulation: current views and future prospects. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220160. [PMID: 37122224 PMCID: PMC10150219 DOI: 10.1098/rstb.2022.0160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/10/2023] [Indexed: 05/02/2023] Open
Abstract
Normal and abnormal cardiac rhythms are of key physiological and clinical interest. This introductory article begins from Sylvio Weidmann's key historic 1950s microelectrode measurements of cardiac electrophysiological activity and Singh & Vaughan Williams's classification of cardiotropic targets. It then proceeds to introduce the insights into cardiomyocyte function and its regulation that subsequently emerged and their therapeutic implications. We recapitulate the resulting view that surface membrane electrophysiological events underlying cardiac excitation and its initiation, conduction and recovery constitute the final common path for the cellular mechanisms that impinge upon this normal or abnormal cardiac electrophysiological activity. We then consider progress in the more recently characterized successive regulatory hierarchies involving Ca2+ homeostasis, excitation-contraction coupling and autonomic G-protein signalling and their often reciprocal interactions with the surface membrane events, and their circadian rhythms. Then follow accounts of longer-term upstream modulation processes involving altered channel expression, cardiomyocyte energetics and hypertrophic and fibrotic cardiac remodelling. Consideration of these developments introduces each of the articles in this Phil. Trans. B theme issue. The findings contained in these articles translate naturally into recent classifications of cardiac electrophysiological targets and drug actions, thereby encouraging future iterations of experimental cardiac electrophysiological discovery, and testing directed towards clinical management. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Christopher L.-H. Huang
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1QW, UK
| | - Ming Lei
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| |
Collapse
|
13
|
Liu Y, Yu M, Wu Y, Wu F, Feng X, Zhao H. Myeloperoxidase in the pericardial fluid improves the performance of prediction rules for postoperative atrial fibrillation. J Thorac Cardiovasc Surg 2023; 165:1064-1077.e8. [PMID: 34275621 DOI: 10.1016/j.jtcvs.2021.06.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 05/15/2021] [Accepted: 06/09/2021] [Indexed: 11/16/2022]
Abstract
OBJECTIVES After surgery, inflammation is a prominent factor influencing postoperative atrial fibrillation. Myeloperoxidase is a major contributor to inflammatory responses after surgical tissue damage. We evaluated whether myeloperoxidase is associated with postoperative atrial fibrillation clinically and in an animal model. METHODS This prospective cohort study included patients undergoing isolated coronary artery bypass grafting. Myeloperoxidase concentrations in blood and pericardial fluid were determined at baseline and 6, 12, and 18 hours after coronary artery bypass grafting. Myeloperoxidase activity in blood, pericardial fluid, and atrium were also evaluated in a canine coronary artery bypass grafting model. Electrophysiologic, histologic, and immunohistochemistry analyses were performed to explore underlying mechanisms. RESULTS Postoperative atrial fibrillation occurred in 45 of 137 patients (32.8%). Patients with postoperative atrial fibrillation had significantly higher serum and pericardial myeloperoxidase levels. Individual clinical and surgical factors had moderate predictive value (area under the curve, 0.760) for postoperative atrial fibrillation. Discrimination improved remarkably when myeloperoxidase was combined with other parameters (area under the curve, 0.901). Pericardial myeloperoxidase at 6 hours postoperatively was the strongest independent predictor of postoperative atrial fibrillation (odds ratio, 19.215). The rate of postoperative atrial fibrillation increased exponentially across pericardial myeloperoxidase grades. Compared with controls, coronary artery bypass grafting-treated dogs showed higher atrial fibrillation vulnerability and maintenance, shorter atrial effective refractory period, attenuated connexin 43 expression, and increased myocardial and pericardial myeloperoxidase activity. Connexin 43 expression and atrial effective refractory period were strongly negatively correlated with myocardial and pericardial myeloperoxidase activity. CONCLUSIONS Myeloperoxidase is linked to postoperative atrial fibrillation, and the ability to predict postoperative atrial fibrillation was remarkably improved by adding pericardial myeloperoxidase. Myeloperoxidase-related atrial structural and electrical remodeling is a physiologic substrate for this arrhythmia.
Collapse
Affiliation(s)
- Yisi Liu
- Capital Medical University, Beijing, P. R. China
| | - Meng Yu
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, P.R. China
| | - Ying Wu
- Capital Medical University, Beijing, P. R. China.
| | - Fangqin Wu
- Capital Medical University, Beijing, P. R. China
| | - Xinwei Feng
- Capital Medical University, Beijing, P. R. China
| | - Haibo Zhao
- Beijing Chao-yang Hospital affiliated to Capital Medical University, Beijing, P.R. China
| |
Collapse
|
14
|
Yang X, Zhao S, Wang S, Cao X, Xu Y, Yan M, Pang M, Yi F, Wang H. Systemic inflammation indicators and risk of incident arrhythmias in 478,524 individuals: evidence from the UK Biobank cohort. BMC Med 2023; 21:76. [PMID: 36855116 PMCID: PMC9976398 DOI: 10.1186/s12916-023-02770-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 02/03/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND The role of systemic inflammation in promoting cardiovascular diseases has attracted attention, but its correlation with various arrhythmias remains to be clarified. We aimed to comprehensively assess the association between various indicators of systemic inflammation and atrial fibrillation/flutter (AF), ventricular arrhythmia (VA), and bradyarrhythmia in the UK Biobank cohort. METHODS After excluding ineligible participants, a total of 478,524 eligible individuals (46.75% male, aged 40-69 years) were enrolled in the study to assess the association between systemic inflammatory indicators and each type of arrhythmia. RESULTS After covariates were fully adjusted, CRP levels were found to have an essentially linear positive correlation with the risk of various arrhythmias; neutrophil count, monocyte count, and NLR showed a non-linear positive correlation; and lymphocyte count, SII, PLR, and LMR showed a U-shaped association. VA showed the strongest association with systemic inflammation indicators, and it was followed sequentially by AF and bradyarrhythmia. CONCLUSIONS Multiple systemic inflammatory indicators showed strong associations with the onset of AF, VA, and bradyarrhythmia, of which the latter two have been rarely studied. Active systemic inflammation management might have favorable effects in reducing the arrhythmia burden and further randomized controlled studies are needed.
Collapse
Affiliation(s)
- Xiaorong Yang
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China.,Clinical Research Center of Shandong University, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shaohua Zhao
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Shaohua Wang
- Department of Internal Medicine, Jinan Hospital, Jinan, China
| | - Xuelei Cao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yue Xu
- Qilu Hospital of Shandong University, Jinan, China
| | - Meichen Yan
- Qilu Hospital of Shandong University, Jinan, China
| | - Mingmin Pang
- Qilu Hospital of Shandong University, Jinan, China
| | - Fan Yi
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China.
| | - Hao Wang
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
15
|
Lage R, Cebro-Márquez M, Vilar-Sánchez ME, González-Melchor L, García-Seara J, Martínez-Sande JL, Fernández-López XA, Aragón-Herrera A, Martínez-Monzonís MA, González-Juanatey JR, Rodríguez-Mañero M, Moscoso I. Circulating miR-451a Expression May Predict Recurrence in Atrial Fibrillation Patients after Catheter Pulmonary Vein Ablation. Cells 2023; 12:cells12040638. [PMID: 36831306 PMCID: PMC9953933 DOI: 10.3390/cells12040638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/07/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Atrial fibrillation is the most prevalent tachyarrhythmia in clinical practice, with very high cardiovascular morbidity and mortality with a high-cost impact in health systems. Currently, it is one of the main causes of stroke and subsequent heart failure and sudden death. miRNAs mediate in several processes involved in cardiovascular disease, including fibrosis and electrical and structural remodeling. Several studies suggest a key role of miRNAs in the course and maintenance of atrial fibrillation. In our study, we aimed to identify the differential expression of circulating miRNAs and their predictive value as biomarkers of recurrence in atrial fibrillation patients undergoing catheter pulmonary vein ablation. To this effect, 42 atrial fibrillation patients were recruited for catheter ablation. We measured the expression of 84 miRNAs in non-recurrent and recurrent groups (45.2%), both in plasma from peripheral and left atrium blood. Expression analysis showed that miRNA-451a is downregulated in recurrent patients. Receiver operating characteristic curve analysis showed that miR-451a in left atrium plasma could predict atrial fibrillation recurrence after pulmonary vein isolation. In addition, atrial fibrillation recurrence is positively associated with the increment of scar percentage. Our data suggest that miRNA-451a expression plays an important role in AF recurrence by controlling fibrosis and progression.
Collapse
Affiliation(s)
- Ricardo Lage
- Cardiology Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - María Cebro-Márquez
- Cardiology Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Marta E. Vilar-Sánchez
- Cardiology Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Laila González-Melchor
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Javier García-Seara
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - José Luis Martínez-Sande
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Xesús Alberte Fernández-López
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Alana Aragón-Herrera
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - María Amparo Martínez-Monzonís
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - José Ramón González-Juanatey
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Moisés Rodríguez-Mañero
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Correspondence: (M.R.-M.); (I.M.); Tel.: +0034-88181-5409 (I.M.)
| | - Isabel Moscoso
- Cardiology Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Correspondence: (M.R.-M.); (I.M.); Tel.: +0034-88181-5409 (I.M.)
| |
Collapse
|
16
|
Matrix Metalloproteinases in Cardioembolic Stroke: From Background to Complications. Int J Mol Sci 2023; 24:ijms24043628. [PMID: 36835040 PMCID: PMC9959608 DOI: 10.3390/ijms24043628] [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: 11/30/2022] [Revised: 01/20/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are endopeptidases participating in physiological processes of the brain, maintaining the blood-brain barrier integrity and playing a critical role in cerebral ischemia. In the acute phase of stroke activity, the expression of MMPs increase and is associated with adverse effects, but in the post-stroke phase, MMPs contribute to the process of healing by remodeling tissue lesions. The imbalance between MMPs and their inhibitors results in excessive fibrosis associated with the enhanced risk of atrial fibrillation (AF), which is the main cause of cardioembolic strokes. MMPs activity disturbances were observed in the development of hypertension, diabetes, heart failure and vascular disease enclosed in CHA2DS2VASc score, the scale commonly used to evaluate the risk of thromboembolic complications risk in AF patients. MMPs involved in hemorrhagic complications of stroke and activated by reperfusion therapy may also worsen the stroke outcome. In the present review, we briefly summarize the role of MMPs in the ischemic stroke with particular consideration of the cardioembolic stroke and its complications. Moreover, we discuss the genetic background, regulation pathways, clinical risk factors and impact of MMPs on the clinical outcome.
Collapse
|
17
|
Parrilla Hernández S, Franck T, Munaut C, Feyereisen É, Piret J, Farnir F, Reigner F, Barrière P, Deleuze S. Characterization of Myeloperoxidase in the Healthy Equine Endometrium. Animals (Basel) 2023; 13:ani13030375. [PMID: 36766264 PMCID: PMC9913682 DOI: 10.3390/ani13030375] [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: 12/14/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Myeloperoxidase (MPO), as a marker of neutrophil activation, has been associated with equine endometritis. However, in absence of inflammation, MPO is constantly detected in the uterine lumen of estrous mares. The aim of this study was to characterize MPO in the uterus of mares under physiological conditions as a first step to better understand the role of this enzyme in equine reproduction. Total and active MPO concentrations were determined, by ELISA and SIEFED assay, respectively, in low-volume lavages from mares in estrus (n = 26), diestrus (n = 18) and anestrus (n = 8) in absence of endometritis. Immunohistochemical analysis was performed on 21 endometrial biopsies randomly selected: estrus (n = 11), diestrus (n = 6) and anestrus (n = 4). MPO, although mostly enzymatically inactive, was present in highly variable concentrations in uterine lavages in all studied phases, with elevated concentrations in estrus and anestrus, while in diestrus, concentrations were much lower. Intracytoplasmic immunoexpression of MPO was detected in the endometrial epithelial cells, neutrophils and glandular secretions. Maximal expression was observed during estrus in mid and basal glands with a predominant intracytoplasmic apical reinforcement. In diestrus, immunopositive glands were sporadic. In anestrus, only the luminal epithelium showed residual MPO immunostaining. These results confirm a constant presence of MPO in the uterine lumen of mares in absence of inflammation, probably as part of the uterine mucosal immune system, and suggest that endometrial cells are a source of uterine MPO under physiological cyclic conditions.
Collapse
Affiliation(s)
- Sonia Parrilla Hernández
- Physiology of Reproduction, Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Thierry Franck
- Center for Oxygen Research and development (CORD), University of Liège, 4000 Liège, Belgium
| | - Carine Munaut
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, 4000 Liège, Belgium
| | - Émilie Feyereisen
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, 4000 Liège, Belgium
| | - Joëlle Piret
- Department of Morphology and Pathology, Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Frédéric Farnir
- Biostatistics and Bioinformatics Applied to Veterinary Sciences, Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | | | | | - Stéfan Deleuze
- Physiology of Reproduction, Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
- Equine and Companion Animal Reproduction, Veterinary Medicine Faculty, University of Liège, 4000 Liège, Belgium
- Correspondence:
| |
Collapse
|
18
|
Tian Y, Liu S, Zhang Y, Yang J, Guo P, Zhang H, Yu X, Zou T. Immune infiltration and immunophenotyping in atrial fibrillation. Aging (Albany NY) 2023; 15:213-229. [PMID: 36602538 PMCID: PMC9876632 DOI: 10.18632/aging.204470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 12/20/2022] [Indexed: 01/05/2023]
Abstract
Atrial fibrillation (AF) is a relatively common arrhythmia in clinical practice. Although significant progress has been achieved in the treatment of AF and its associated complications, research on AF prevention lags behind, mainly due to the lack of a deep understanding of AF pathogenesis. In recent years, as our knowledge has grown, the role of the inflammatory/immune response in the occurrence and progression of AF has gradually gained attention. In this paper, based on existing gene expression data in the Gene Expression Omnibus database, a detailed description of immune infiltration status in AF is presented using a series of analytical methods, including differential analysis, Gene Ontology categorization, Kyoto Encyclopedia of Genes and Genomes enrichment analysis, and weighted gene coexpression network analysis, and analysis tools such as CIBERSORTx and Cytoscape. Several new AF/immune infiltrations-related signature genes were identified, and the AF/immune infiltration pathology was classified based on these immune signature genes, thus providing novel insights into the pathogenesis of AF based on the inflammatory response.
Collapse
Affiliation(s)
- Yuqing Tian
- Department of Cardiology, Affiliated Hospital of Panzhihua University, Panzhihua 617000, Sichuan, P.R. China
- Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Shiying Liu
- Department of Plastic Surgery, Affiliated Hospital of Panzhihua University, Panzhihua 617000, Sichuan, P.R. China
| | - Yanan Zhang
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, P.R. China
| | - Jiefu Yang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing 100730, P.R. China
| | - Peiyao Guo
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing 100730, P.R. China
- Peking University Fifth School of Clinical Medicine, Beijing Hospital, Beijing 100730, P.R. China
| | - Hongchao Zhang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing 100730, P.R. China
| | - Xue Yu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing 100730, P.R. China
| | - Tong Zou
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing 100730, P.R. China
| |
Collapse
|
19
|
Wang J, Hu S, Liang C, Ling Y. The association between systemic inflammatory response index and new-onset atrial fibrillation in patients with ST-elevated myocardial infarction treated with percutaneous coronary intervention. BMC Cardiovasc Disord 2022; 22:525. [PMID: 36474135 PMCID: PMC9724303 DOI: 10.1186/s12872-022-02989-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND New-onset atrial fibrillation (NOAF) complicating with ST-elevated myocardial infarction (STEMI) patients following percutaneous coronary intervention (PCI) is associated with worse prognosis. The systemic inflammatory response index (SIRI), serves as a novel inflammatory indicator, is found to be predictive of adverse outcomes. The aim of this study is to explore the association between NOAF and SIRI. METHODS A retrospective data included 616 STEMI participants treated with PCI in our cardiology department had been analyzed in present investigation, of which being divided into a NOAF or sinus rhythm (SR) group based on the presence or absence of atrial fibrillation. The predictive role of SIRI for in detecting NOAF had been evaluated by the logistic regression analyses and receiver operating characteristic (ROC) curve. Additionally, long-term all-cause mortality between both groups was compared using the Kaplan-Meier test. RESULTS NOAF during hospitalization developed in 7.6% of PCI-treated individuals. After multivariate regression analyses, SIRI remains to be an independently predictor of NOAF (odds ratio 1.782, 95% confidence interval 1.675-1.906, P = 0.001). In the ROC curve analysis, SIRI with a cut-off value of 4.86 was calculated to predict NOAF, with 4.86, with a sensitivity of 80.85% and a specificity of 75.57%, respectively (area under the curve (AUC) = 0.826, P < 0.001). Furthermore, pairwise compassion of ROC curves displayed the superiority of SIRI in the prediction of NOAF in comparison with that of neutrophil/lymphocyte or monocyte/lymphocyte (P < 0.05). In addition, the participants in NOAF group had a significantly higher incidence of all-cause death compared to those in SR group after a median of 40-month follow-up (22.0% vs 5.8%, log-rank P < 0.001). CONCLUSION SIRI can independently predict NOAF in patients with STEMI after PCI, with being positively correlated to worsened outcomes.
Collapse
Affiliation(s)
- Jingfeng Wang
- grid.443626.10000 0004 1798 4069Department of Cardiology, Yijishan Hospital Affiliated to Wannan Medical College, 2# West Zhe Shan Road, Wuhu, 241000 China
| | - Sisi Hu
- grid.443626.10000 0004 1798 4069Department of Cardiology, Yijishan Hospital Affiliated to Wannan Medical College, 2# West Zhe Shan Road, Wuhu, 241000 China
| | - Cheng Liang
- grid.443626.10000 0004 1798 4069Department of Cardiology, Yijishan Hospital Affiliated to Wannan Medical College, 2# West Zhe Shan Road, Wuhu, 241000 China
| | - Yang Ling
- grid.443626.10000 0004 1798 4069Department of Cardiology, Yijishan Hospital Affiliated to Wannan Medical College, 2# West Zhe Shan Road, Wuhu, 241000 China
| |
Collapse
|
20
|
Sun W, Li H, Wang Z, Li Q, Wen H, Wu Y, Du J. Elevated tissue inhibitor of metalloproteinase-1 along with left atrium hypertrophy predict atrial fibrillation recurrence after catheter ablation. Front Cardiovasc Med 2022; 9:1010443. [PMID: 36386356 PMCID: PMC9663807 DOI: 10.3389/fcvm.2022.1010443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
This study aimed to establish a model that predicts atrial fibrillation (AF) recurrence after catheter ablation using clinical risk factors and biomarkers. We used a prospective cohort study, including 230 consecutive persistent AF patients successfully treated with catheter ablation from January 2019 to December 2020 in our hospital. AF recurrence was followed-up after catheter ablation, and clinical risk factors and biomarkers for AF recurrence were analyzed. AF recurred after radiofrequency ablation in 72 (31%) patients. Multiple multivariate logistic regression analysis demonstrated that tissue inhibitor of metalloproteinase-1 (TIMP-1) and left atrium diameter (LAd) were closely associated with AF recurrence. The prediction model constructed by combining TIMP-1 and LAd effectively predicted AF recurrence. Additionally, the model’s performance discrimination, accuracy, and calibration were confirmed through internal validation using bootstrap resampling (1,000 times). The model showed good fitting (Hosmer–Lemeshow goodness chi-square 3.76138, p = 0.926) and had a superior discrimination ability (the area under the receiver operation characteristic curve0.917; 95% CI 0.882–0.952). The calibration curve showed good agreement between the predicted probability and the actual probability. Moreover, the decision curve analysis (DCA) showed the clinical useful of the nomogram. In conclusion, our predictive model based on serum TIMP-1 and LAd levels could predict AF recurrence after catheter ablation.
Collapse
Affiliation(s)
- Weiping Sun
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| | - Haiwei Li
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zefeng Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Qin Li
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| | - Haichu Wen
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| | - Yongquan Wu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yongquan Wu,
| | - Jie Du
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
- Jie Du,
| |
Collapse
|
21
|
Li Y, Xia Q, Zhu C, Cao W, Xia Z, Liu X, Xiao B, Chen K, Liu Y, Zhong L, Tan B, Lei J, Zhu J. An activatable Mn(II) MRI probe for detecting peroxidase activity in vitro and in vivo. J Inorg Biochem 2022; 236:111979. [PMID: 36087435 DOI: 10.1016/j.jinorgbio.2022.111979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 12/15/2022]
Abstract
Myeloperoxidase (MPO), a hallmark of the function and activation of innate immune cells, can act as a 'double-edged sword', contributing to clear infection as well as causing tissue oxidizing damage in various inflammatory diseases. In this study, an activatable Mn(II) chelate-based magnetic resonance imaging (MRI) contrast agent (CA), Mn-TyEDTA (TyEDTA = tyrosine derived ethylenediaminetetraacetic acid) structurally featuring a phenol group as the electron-donor, was developed to sense the activity of peroxidase in vitro and in vivo. Mn-TyEDTA demonstrated a peroxidase activity-dependent relaxivity in the presence of horseradish peroxidase (HRP)/H2O2 with more than a 2.6-fold increase in water proton relaxivity produced (HRP, 500 U; H2O2, 4.5 eq). A mechanism of peroxidase-mediated Mn(II) monomer radical polymerization was confirmed with those oligomers of Mn-TyEDTA such as dimer, trimer and tetramer were found in the LC-MS study. Dynamic MR imaging of normal mice revealed rapid blood clearance and mixed renal and hepatobiliary elimination of Mn-TyEDTA. Furthermore, compared to liver-specific and non-specific extracellular contrast agents (Mn-BnO-TyEDTA (BnO-TyEDTA = benzyl tyrosine-derived ethylenediaminetetraacetic acid) and Gd-DTPA (DTPA = diethylene triamine penta-acetic acid)), MRI on a monosodium urate (MSU) crystal-induced acute mice model of arthritis showed that inflamed tissues could be selectively enhanced by Mn-TyEDTA, suggesting that this peroxidase-activatable Mn(II) MRI probe could potentially be used for noninvasive detection of MPO activity in vivo.
Collapse
Affiliation(s)
- Yunhe Li
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China; School of Pharmacy, North Sichuan Medical College, Fujiang Road 234, Nanchong City, Sichuan 637000, China
| | - Qian Xia
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China
| | - Chunrong Zhu
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China
| | - Weidong Cao
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China; School of Pharmacy, North Sichuan Medical College, Fujiang Road 234, Nanchong City, Sichuan 637000, China
| | - Zhiyang Xia
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China
| | - Xinxin Liu
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China
| | - Bin Xiao
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China
| | - Keyu Chen
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China; School of Pharmacy, North Sichuan Medical College, Fujiang Road 234, Nanchong City, Sichuan 637000, China
| | - Yun Liu
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China
| | - Lei Zhong
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China
| | - Bangxian Tan
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China
| | - Jun Lei
- School of Pharmacy, North Sichuan Medical College, Fujiang Road 234, Nanchong City, Sichuan 637000, China.
| | - Jiang Zhu
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China; School of Pharmacy, North Sichuan Medical College, Fujiang Road 234, Nanchong City, Sichuan 637000, China.
| |
Collapse
|
22
|
Markers of Inflammation, Oxidative Stress, and Fibrosis in Patients with Atrial Fibrillation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4556671. [PMID: 35651726 PMCID: PMC9150993 DOI: 10.1155/2022/4556671] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 05/01/2022] [Accepted: 05/05/2022] [Indexed: 11/17/2022]
Abstract
Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia in clinical practice. The pathogenesis of AF is linked to inflammatory reaction and oxidative stress, which leads to fibrosis of the atria and progression of the disease. The purpose of this study was to define the role of several biomarkers of inflammation, fibrosis, and oxidative stress (OxS). We included 75 patients with paroxysmal/persistent AF, who were admitted for electrical cardioversion or pulmonary vein isolation (PVI). High-sensitivity C-reactive protein (hsCRP), galectin-3 (Gal-3), myeloperoxidase (MPO), oxidized low-density lipoprotein (oxLDL), and N-terminal pro-brain natriuretic peptide (NT-proBNP) were measured before the procedures. We compared the results with those of 75 healthy age-, sex-, and blood pressure-matched individuals. The patients were followed up for 1 year after the intervention to establish the recurrence of AF and its association with the measured markers. Patients with AF had higher MPO (52.6 vs. 36.2 ng/ml, p < 0.001) and NT-proBNP (209.0 vs. 28.0 pg/ml, p < 0.001) compared to healthy subjects. Also, they showed significantly higher levels of hsCRP (1.5 vs. 1.1 mg/l, p = 0.001) and Gal-3 (11.4 vs. 9.7 mg/l, p = 0.003), while there was no difference found in oxLDL (71.5 vs. 71.7 U/l, p = 0.449). MPO (OR = 1.012, p = 0.014), hsCRP (OR = 1.265, p = 0.026), and weight (OR = 1.029, p = 0.013) were independently associated with AF in a multivariable logistic regression analysis. Patients with successful maintenance of sinus rhythm (SR) for one year had lower baseline MPO (40.5 vs. 84.3 ng/ml, p = 0.005) and NT-proBNP (127.5 vs. 694.0 pg/ml, p < 0.001) compared to patients with recurrent AF episodes, but there was no difference in hsCRP, Gal-3, or oxLDL between them. MPO (OR = 0.985, p = 0.010) was independently associated with AF recurrence during the follow-up period when adjusted for cofounders. Patients with AF had increased markers of inflammation and fibrosis, while there was no increase detected in the OxS marker oxLDL. MPO was independently associated with AF in a multivariate model. Inflammatory and fibrotic mechanisms are important factors in electrical and structural remodelling progress in the atria of patients with AF.
Collapse
|
23
|
Fei J, Demillard LJ, Ren J. Reactive oxygen species in cardiovascular diseases: an update. EXPLORATION OF MEDICINE 2022. [DOI: 10.37349/emed.2022.00085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Cardiovascular diseases are among the leading causes of death worldwide, imposing major health threats. Reactive oxygen species (ROS) are one of the most important products from the process of redox reactions. In the onset and progression of cardiovascular diseases, ROS are believed to heavily influence homeostasis of lipids, proteins, DNA, mitochondria, and energy metabolism. As ROS production increases, the heart is damaged, leading to further production of ROS. The vicious cycle continues on as additional ROS are generated. For example, recent evidence indicated that connexin 43 (Cx43) deficiency and pyruvate kinase M2 (PKM2) activation led to a loss of protection in cardiomyocytes. In this context, a better understanding of the mechanisms behind ROS production is vital in determining effective treatment and management strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Juanjuan Fei
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Laurie J. Demillard
- School of Pharmacy, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
24
|
Training associated alterations in equine respiratory immunity using a multiomics comparative approach. Sci Rep 2022; 12:427. [PMID: 35013475 PMCID: PMC8748960 DOI: 10.1038/s41598-021-04137-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/07/2021] [Indexed: 12/19/2022] Open
Abstract
Neutrophilic airway inflammation is highly prevalent in racehorses in training, with the term mild to moderate equine asthma (MMEA) being applied to the majority of such cases. Our proposed study is largely derived from the strong association between MMEA in racehorses and their entry into a race training program. The objectives of this study are to characterise the effect of training on the local pulmonary immune system by defining the gene and protein expression of tracheal wash (TW) derived samples from Thoroughbred racehorses prior to and following commencement of race training. Multiomics analysis detected 2138 differentially expressed genes and 260 proteins during the training period. Gene and protein sets were enriched for biological processes related to acute phase response, oxidative stress, haemopoietic processes, as well as to immune response and inflammation. This study demonstrated TW samples to represent a rich source of airway cells, protein and RNA to study airway immunity in the horse and highlighted the benefits of a multiomics methodological approach to studying the dynamics of equine airway immunity. Findings likely reflect the known associations between race-training and both airway inflammation and bleeding, offering further insight into the potential mechanisms which underpin training associated airway inflammation.
Collapse
|
25
|
Cardiac fibrosis and atrial fibrillation. POSTEP HIG MED DOSW 2022. [DOI: 10.2478/ahem-2022-0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Cardiac fibrosis is characterized by the imbalance of production and degradation of the extracellular matrix. The result of this process is an accumulation of scar tissue, which is associated with many pathological processes such as excessive mechanical stress on the heart, inflammation, ischemia, oxidative stress, or excessive neurohormonal activation. Fibrotic response results in damaged heart architecture and dysfunction of the heart. Cardiac fibrosis leads to increased stiffness of the left ventricle and arteries, promotes disorders of contraction and relaxation of the heart, disrupts electrophysiology of heart cells, and induces arrhythmias.
Atrial fibrillation is one of the most common arrhythmias. It is associated with a deterioration in the quality of life and more frequent use of medical assistance. It is also an instantaneous risk factor for many diseases, including stroke. The underlying cause of this arrhythmia is electrical and structural remodeling induced by cardiac fibrosis. Therefore, much attention is paid to the search for biochemical markers that would allow non-invasive determination of the degree of this fibrosis.
The promising markers include galectin-3, human epididymis protein 4 (HE4), serum soluble ST2, and adipose triglyceride lipase (ATGL). Studies have shown that plasma concentrations of these substances reflect the degree of myocardial fibrosis and are indirectly associated with AF.
There are high hopes for the use of these markers in patients undergoing arrhythmia ablation. More research is needed to confirm that these markers can be used to estimate the chance of maintaining sinus rhythm in patients after ablation.
Collapse
|
26
|
Liu C, Ma N, Guo Z, Zhang Y, Zhang J, Yang F, Su X, Zhang G, Xiong X, Xing Y. Relevance of mitochondrial oxidative stress to arrhythmias: Innovative concepts to target treatments. Pharmacol Res 2021; 175:106027. [PMID: 34890774 DOI: 10.1016/j.phrs.2021.106027] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/26/2021] [Accepted: 12/05/2021] [Indexed: 12/13/2022]
Abstract
Cardiac arrhythmia occurs frequently worldwide, and in severe cases can be fatal. Mitochondria are the power plants of cardiomyocytes. In recent studies, mitochondria under certain stimuli produced excessive reactive oxygen species (ROS), which affect the normal function of cardiomyocytes through ion channels and related proteins. Mitochondrial oxidative stress (MOS) plays a key role in diseases with multifactorial etiopathogenesis, such as arrhythmia; MOS can lead to arrhythmias such as atrial fibrillation and ventricular tachycardia. This review discusses the mechanisms of arrhythmias caused by MOS, particularly of ROS produced by mitochondria. MOS can cause arrhythmias by affecting the activities of Ca2+-related proteins, the mitochondrial permeability transition pore protein, connexin 43, hyperpolarization-activated cyclic nucleotide-gated potassium channel 4, and ion channels. Based on these mechanisms, we discuss possible new treatments for arrhythmia. Targeted treatments focusing on mitochondria may reduce the progression of arrhythmias, as well as the occurrence of severe arrhythmias, and may be effective for personalized disease prevention.
Collapse
Affiliation(s)
- Can Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ning Ma
- Dezhou Second People's Hospital, Dezhou 253000, China
| | - Ziru Guo
- Xingtai People's Hospital, Xingtai 054001, China
| | - Yijun Zhang
- The First Affiliated Hospital, Hebei North University, Zhangjiakou 075000, China
| | - Jianzhen Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Fan Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xin Su
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Guoxia Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xingjiang Xiong
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Yanwei Xing
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
27
|
Leiva O, AbdelHameid D, Connors JM, Cannon CP, Bhatt DL. Common Pathophysiology in Cancer, Atrial Fibrillation, Atherosclerosis, and Thrombosis: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2021; 3:619-634. [PMID: 34988471 PMCID: PMC8702799 DOI: 10.1016/j.jaccao.2021.08.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease and cancer are the 2 leading causes of death worldwide. Emerging evidence suggests common mechanisms between cancer and cardiovascular disease, including atrial fibrillation and atherosclerosis. With advances in cancer therapies, screening, and diagnostics, cancer-specific survival and outcomes have improved. This increase in survival has led to the coincidence of cardiovascular disease, including atrial fibrillation and atherosclerosis, as patients with cancer live longer. Additionally, cancer and cardiovascular disease share several risk factors and underlying pathophysiologic mechanisms, including inflammation, cancer-related factors including treatment effects, and alterations in platelet function. Patients with cancer are at increased risk for bleeding and thrombosis compared with the general population. Although optimal antithrombotic therapy, including agent choice and duration, has been extensively studied in the general population, this area remains understudied in patients with cancer despite their altered thrombotic and bleeding risk. Future investigation, including incorporation of cancer-specific characteristics to traditional thrombotic and bleeding risk scores, clinical trials in the cancer population, and the development of novel antithrombotic and anti-inflammatory strategies on the basis of shared pathophysiologic mechanisms, is warranted to improve outcomes in this patient population.
Collapse
Key Words
- AF, atrial fibrillation
- CAD, coronary artery disease
- CHIP, clonal hematopoiesis of indeterminate potential
- CI, confidence interval
- CLEC-2, C-type lectin-like receptor 2
- HR, hazard ratio
- IL, interleukin
- MI, myocardial infarction
- PCI, percutaneous coronary intervention
- ROS, reactive oxygen species
- TKI, tyrosine kinase inhibitor
- VTE, venous thromboembolism
- arrhythmia
- risk factor
- thrombosis
Collapse
Affiliation(s)
- Orly Leiva
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Duaa AbdelHameid
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jean M. Connors
- Division of Hematology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Christopher P. Cannon
- Brigham and Women’s Hospital Heart & Vascular Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Deepak L. Bhatt
- Brigham and Women’s Hospital Heart & Vascular Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
28
|
Yan T, Zhu S, Shi Y, Xie C, Zhu M, Zhang Y, Wang C, Guo C. Pan-Cancer Analysis of Atrial-Fibrillation-Related Innate Immunity Gene ANXA4. Front Cardiovasc Med 2021; 8:713983. [PMID: 34540918 PMCID: PMC8446278 DOI: 10.3389/fcvm.2021.713983] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/16/2021] [Indexed: 01/11/2023] Open
Abstract
Background: Atrial fibrillation (AF) is the most common tachyarrhythmia around the world. Cancer is one of the main causes of death worldwide. A recent study demonstrated that cancer was associated with an increased incidence of AF. In the present study, we aimed to explore possible mechanisms and potential common therapeutic targets between AF and cancer. Methods: Differentially expressed proteins between AF and sinus rhythm were identified utilizing proteomics analysis. Weighted gene correlation network analysis was applied to cluster proteins into different modules and investigate associations between modules and AF. Hub immune-related genes were selected via InnateDB database and verified using qRT-PCR. RNA sequencing and clinical data of 33 different cancer types were achieved from The Cancer Genome Atlas (TCGA). The correlations between ANXA4 expression and the prognosis were calculated utilizing Cox regression analysis and Kaplan-Meier survival analysis. Spearman's rank correlation test was used to assess associations between ANXA4 and immune infiltration and DNA methylation. Enrichment analysis was performed through gene ontology and gene set enrichment analysis (GSEA). Results:ANXA4 was identified as hub immune-related gene between AF and sinus rhythm. Expression levels of ANXA4 increased in diverse cancer types. Survival analysis suggested prognostic significance of ANXA4 expression levels in various cancer types. Immune correlation analysis indicated that ANXA4 expression levels were associated with tumor immune infiltration in most cancer types. ANXA4 might influence the efficacy of immunotherapy via tumor burden and microsatellite instability. GSEA results indicated that high ANXA4 expression groups were mainly enriched in peroxisome, bile acid biosynthesis, and p53 pathway. Conclusion:ANXA4 was identified as a hub immune-related gene in AF, which has never been reported. Pan-cancer analysis indicated its potential as a novel clinical prognostic marker and therapeutic target in diverse cancer types. ANXA4 might play crucial roles in AF and cancer, and targeted therapy for ANXA4 might reduce the incidence of AF in cancer patients.
Collapse
Affiliation(s)
- Tao Yan
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shijie Zhu
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Shi
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Changming Xie
- Department of Cardiovascular, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Miao Zhu
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yangyang Zhang
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiovascular Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Chunsheng Wang
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Changfa Guo
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Xintarakou A, Tzeis S, Psarras S, Asvestas D, Vardas P. Atrial fibrosis as a dominant factor for the development of atrial fibrillation: facts and gaps. Europace 2021; 22:342-351. [PMID: 31998939 DOI: 10.1093/europace/euaa009] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/03/2020] [Indexed: 01/08/2023] Open
Abstract
Atrial fibrillation (AF), the most commonly diagnosed arrhythmia, affects a notable percentage of the population and constitutes a major risk factor for thromboembolic events and other heart-related conditions. Fibrosis plays an important role in the onset and perpetuation of AF through structural and electrical remodelling processes. Multiple molecular pathways are involved in atrial substrate modification and the subsequent maintenance of AF. In this review, we aim to recapitulate underlying molecular pathways leading to atrial fibrosis and to indicate existing gaps in the complex interplay of atrial fibrosis and AF.
Collapse
Affiliation(s)
| | - Stylianos Tzeis
- Cardiology Department, Mitera General Hospital, Hygeia Group, Athens, Greece
| | - Stelios Psarras
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Greece
| | - Dimitrios Asvestas
- Cardiology Department, Mitera General Hospital, Hygeia Group, Athens, Greece
| | - Panos Vardas
- Heart Sector, Hygeia Hospitals Group, 5, Erithrou Stavrou, Marousi, Athens 15123, Greece
| |
Collapse
|
30
|
Diteepeng T, Del Monte F, Luciani M. The long and winding road to target protein misfolding in cardiovascular diseases. Eur J Clin Invest 2021; 51:e13504. [PMID: 33527342 DOI: 10.1111/eci.13504] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/18/2021] [Accepted: 01/26/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND In the last decades, cardiovascular diseases (CVD) have remained the first leading cause of mortality and morbidity in the world. Although several therapeutic approaches have been introduced in the past, the development of novel treatments remains an important research goal, which is hampered by the lack of understanding of key mechanisms and targets. Emerging evidences in recent years indicate the involvement of misfolded proteins aggregation and the derailment of protein quality control in the pathogenesis of cardiovascular diseases. Several potential interventions targeting protein quality control have been translated from the bench to the bedside to effectively employ the misfolded proteins as promising therapeutic targets for cardiac diseases, but with trivial results. DESIGN In this review, we describe the recent progresses in preclinical and clinical studies of protein misfolding and compromised protein quality control by selecting and reporting studies focusing on cardiovascular diseases including cardiomyopathies, cardiac amyloidosis, atherosclerosis, atrial fibrillation and thrombosis. RESULTS In preclinical models, modulators of several molecular targets (eg heat shock proteins, unfolded protein response, ubiquitin protein system, autophagy and histone deacetylases) have been tested in various conditions with promising results although lacking an adequate transition towards clinical setting. CONCLUSIONS At present, no therapeutic strategies have been reported to attenuate proteotoxicity in patients with CVD due to a lack of specific biomarkers for pinpointing upstream events in protein folding defects at a subclinical stage of the diseases requiring an intensive collaboration between basic scientists and clinicians.
Collapse
Affiliation(s)
- Thamonwan Diteepeng
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Federica Del Monte
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC, USA.,Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna Alma Mater, Bologna, Italy
| | - Marco Luciani
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.,Department of Internal Medicine, Cantonal Hospital of Baden, Baden, Switzerland
| |
Collapse
|
31
|
Correa S, Curtis KA, Waikar SS, Mc Causland FR. Serum Myeloperoxidase, Uric Acid, and the Risk of Atrial Fibrillation in Chronic Kidney Disease. Circ Arrhythm Electrophysiol 2021; 14:e009483. [PMID: 33858180 DOI: 10.1161/circep.120.009483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Simon Correa
- Division of Renal Medicine, Brigham and Women's Hospital (S.C., K.A.C., F.R.M.C.), Boston University School of Medicine, Boston Medical Center, MA.,Harvard Medical School (S.C., K.A.C., F.R.M.C.), Boston University School of Medicine, Boston Medical Center, MA
| | - Katherine A Curtis
- Division of Renal Medicine, Brigham and Women's Hospital (S.C., K.A.C., F.R.M.C.), Boston University School of Medicine, Boston Medical Center, MA.,Harvard Medical School (S.C., K.A.C., F.R.M.C.), Boston University School of Medicine, Boston Medical Center, MA
| | - Sushrut S Waikar
- Section of Nephrology, Department of Medicine (S.S.W.), Boston University School of Medicine, Boston Medical Center, MA
| | - Finnian R Mc Causland
- Division of Renal Medicine, Brigham and Women's Hospital (S.C., K.A.C., F.R.M.C.), Boston University School of Medicine, Boston Medical Center, MA.,Harvard Medical School (S.C., K.A.C., F.R.M.C.), Boston University School of Medicine, Boston Medical Center, MA
| |
Collapse
|
32
|
Role of Oxidative DNA Damage and Repair in Atrial Fibrillation and Ischemic Heart Disease. Int J Mol Sci 2021; 22:ijms22083838. [PMID: 33917194 PMCID: PMC8068079 DOI: 10.3390/ijms22083838] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023] Open
Abstract
Atrial fibrillation (AF) and ischemic heart disease (IHD) represent the two most common clinical cardiac diseases, characterized by angina, arrhythmia, myocardial damage, and cardiac dysfunction, significantly contributing to cardiovascular morbidity and mortality and posing a heavy socio-economic burden on society worldwide. Current treatments of these two diseases are mainly symptomatic and lack efficacy. There is thus an urgent need to develop novel therapies based on the underlying pathophysiological mechanisms. Emerging evidence indicates that oxidative DNA damage might be a major underlying mechanism that promotes a variety of cardiac diseases, including AF and IHD. Antioxidants, nicotinamide adenine dinucleotide (NAD+) boosters, and enzymes involved in oxidative DNA repair processes have been shown to attenuate oxidative damage to DNA, making them potential therapeutic targets for AF and IHD. In this review, we first summarize the main molecular mechanisms responsible for oxidative DNA damage and repair both in nuclei and mitochondria, then describe the effects of oxidative DNA damage on the development of AF and IHD, and finally discuss potential targets for oxidative DNA repair-based therapeutic approaches for these two cardiac diseases.
Collapse
|
33
|
Wang P, Cheng M, Wang P, Xiong L, Zeng Y, Tu X, Zhang R, Xia Y, Wu G, Wang Q, Cheng X, Xu C. SNP rs2243828 in MPO associated with myeloperoxidase level and atrial fibrillation risk in Chinese Han population. J Cell Mol Med 2021; 24:10263-10266. [PMID: 33460291 PMCID: PMC7520285 DOI: 10.1111/jcmm.15644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 06/10/2020] [Accepted: 06/29/2020] [Indexed: 12/01/2022] Open
Abstract
Previous studies shown that myeloperoxidase (MPO) level is higher in patients with atrial fibrillation (AF); however, no genetic evidence between MPO and AF risk in human population was observed. Therefore, the present study was aimed to investigate the association between rs2243828, a variant in promoter region of MPO and the risk of AF in Chinese GeneID population. The results demonstrated that the minor G allele of rs2243828 showed a significant association with AF in two independent population (GeneID‐north population with 694 AF cases and 710 controls, adjusted P‐adj = 6.25 × 10−3 with an odds ratio was 0.77, GeneID‐central population with 1106 cases and 1501 controls, P‐adj = 9.88 × 10−5 with an odds ratio was 0.75). The results also showed G allele was significantly associated with lower plasma concentration of myeloperoxidase in general population. We also observed a significant difference of odds ratio between subgroups of hypertension and non‐hypertension. Therefore, our findings identified variant in MPO associated with risk of AF and it may give strong evidence to link the inflammation with the incidence of AF.
Collapse
Affiliation(s)
- Pengyun Wang
- Department of Clinical Laboratory, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mian Cheng
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengxia Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research and Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Xiong
- Department of Clinical Laboratory, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yali Zeng
- Department of Clinical Laboratory, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Tu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research and Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, China
| | - Rongfeng Zhang
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yunlong Xia
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Gang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research and Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, China.,Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, OH, USA.,Department of Molecular Medicine, CCLCM, Cleveland, OH, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research and Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
34
|
Myeloperoxidase Inhibition Decreases the Expression of Collagen and Metallopeptidase in Mare Endometria under In Vitro Conditions. Animals (Basel) 2021; 11:ani11010208. [PMID: 33467081 PMCID: PMC7830995 DOI: 10.3390/ani11010208] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/21/2020] [Accepted: 01/14/2021] [Indexed: 12/19/2022] Open
Abstract
Neutrophils can originate neutrophil extracellular traps (NETs). Myeloperoxidase (MPO) is a peroxidase found in NETs associated to equine endometrosis and can be inhibited by 4-aminobenzoic acid hydrazide (ABAH). Metallopeptidases (MMPs) participate in extracellular matrix stability and fibrosis development. The objectives of this in vitro work were to investigate, in explants of mare's endometrium, (i) the ABAH capacity to inhibit MPO-induced collagen type I (COL1) expression; and (ii) the action of MPO and ABAH on the expression and gelatinolytic activity of MMP-2/-9. Explants retrieved from the endometrium of mares in follicular or mid-luteal phases were treated with MPO, ABAH, or their combination, for 24 or 48 h. The qPCR analysis measured the transcription of COL1A2, MMP2, and MMP9. Western blot and zymography were performed to evaluate COL1 protein relative abundance and gelatinolytic activity of MMP-2/-9, respectively. Myeloperoxidase elevated COL1 relative protein abundance at both treatment times in follicular phase (p < 0.05). The capacity of ABAH to inhibit MPO-induced COL1 was detected in follicular phase at 48 h (p < 0.05). The gelatinolytic activity of activated MMP-2 augmented in mid-luteal phase at 24 h after MPO treatment, but it was reduced with MPO+ABAH treatment. The activity of MMP-9 active form augmented in MPO-treated explants. However, this effect was inhibited by ABAH in the follicular phase at 48 h (p < 0.05). By inhibiting the pro-fibrotic effects of MPO, it might be possible to reduce the development of endometrosis. Metallopeptidase-2 might be involved in an acute response to MPO in the mid-luteal phase, while MMP-9 might be implicated in a prolonged exposition to MPO in the follicular phase.
Collapse
|
35
|
Rawat K, Syeda S, Shrivastava A. Neutrophil-derived granule cargoes: paving the way for tumor growth and progression. Cancer Metastasis Rev 2021; 40:221-244. [PMID: 33438104 PMCID: PMC7802614 DOI: 10.1007/s10555-020-09951-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/22/2020] [Indexed: 01/31/2023]
Abstract
Neutrophils are the key cells of our innate immune system mediating host defense via a range of effector functions including phagocytosis, degranulation, and NETosis. For this, they employ an arsenal of anti-microbial cargoes packed in their readily mobilizable granule subsets. Notably, the release of granule content is tightly regulated; however, under certain circumstances, their unregulated release can aggravate tissue damage and could be detrimental to the host. Several constituents of neutrophil granules have also been associated with various inflammatory diseases including cancer. In cancer setting, their excessive release may modulate tissue microenvironment which ultimately leads the way for tumor initiation, growth and metastasis. Neutrophils actively infiltrate within tumor tissues, wherein they show diverse phenotypic and functional heterogeneity. While most studies are focused at understanding the phenotypic heterogeneity of neutrophils, their functional heterogeneity, much of which is likely orchestrated by their granule cargoes, is beginning to emerge. Therefore, a better understanding of neutrophil granules and their cargoes will not only shed light on their diverse role in cancer but will also reveal them as novel therapeutic targets. This review provides an overview on existing knowledge of neutrophil granules and detailed insight into the pathological relevance of their cargoes in cancer. In addition, we also discuss the therapeutic approach for targeting neutrophils or their microenvironment in disease setting that will pave the way forward for future research.
Collapse
Affiliation(s)
- Kavita Rawat
- grid.8195.50000 0001 2109 4999Department of Zoology, University of Delhi, Delhi, 110007 India
| | - Saima Syeda
- grid.8195.50000 0001 2109 4999Department of Zoology, University of Delhi, Delhi, 110007 India
| | - Anju Shrivastava
- grid.8195.50000 0001 2109 4999Department of Zoology, University of Delhi, Delhi, 110007 India
| |
Collapse
|
36
|
He L, Liu R, Yue H, Ren S, Zhu G, Guo Y, Qin C. Actin-granule formation is an additional step in cardiac myofibroblast differentiation. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:165. [PMID: 33569467 PMCID: PMC7867932 DOI: 10.21037/atm-20-8231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background Atrial fibrillation is the most common and long-lasting cardiac arrhythmia, and profoundly effects the daily lives of patients. The pathogenesis and persistence of atrial fibrillation is closely related to the cardiac fibroblast and its myofibroblast differentiation as increased collagen synthesis and migration capability. Thus better understanding of myofibroblast differentiation is essential for the prevention and treatment of atrial fibrillation. Methods Cardiac fibroblasts were isolated from neonatal rats and its actin structure was analyzed by immunofluorescence staining. Myofibroblast differentiation was induced by Angiotensin II (Ang II) and ROCK signaling related proteins were determined by western blot. Fasudil and Ricolinostat were employed to abrogate ROCK signaling and their effects on myofibroblast differentiation were assessed by IF microscopy and Celigo Image Cytometry. Results Stress actin fibers similar to actin filaments in myofibroblast differentiation are regulated by ROCK signaling, and our results also suggested Guanine nucleotide exchange factor-H1 (GEF-H1) phosphorylation could be induced by Ang II. In addition, Fasudil could down-regulate RhoA, GEF-H1, and phosphorylated GEF-H1 to inhibit ROCK signaling and further reduce Col I expression and the myofibroblast proportion. Conclusions An individual phase characterized by actin-granule formation was identified in cardiac myofibroblast differentiation. In the meanwhile, myofibroblast differentiation and its F-actin assembly could be detained in this phase by Fasudil abrogating the ROCK signaling pathway.
Collapse
Affiliation(s)
- Li He
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ruiqi Liu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Honghua Yue
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shuofang Ren
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Guonian Zhu
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, China
| | - Yingqiang Guo
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Chaoyi Qin
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
37
|
Mason FE, Pronto JRD, Alhussini K, Maack C, Voigt N. Cellular and mitochondrial mechanisms of atrial fibrillation. Basic Res Cardiol 2020; 115:72. [PMID: 33258071 PMCID: PMC7704501 DOI: 10.1007/s00395-020-00827-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/26/2020] [Indexed: 11/06/2022]
Abstract
The molecular mechanisms underlying atrial fibrillation (AF), the most common form of arrhythmia, are poorly understood and therefore target-specific treatment options remain an unmet clinical need. Excitation–contraction coupling in cardiac myocytes requires high amounts of adenosine triphosphate (ATP), which is replenished by oxidative phosphorylation in mitochondria. Calcium (Ca2+) is a key regulator of mitochondrial function by stimulating the Krebs cycle, which produces nicotinamide adenine dinucleotide for ATP production at the electron transport chain and nicotinamide adenine dinucleotide phosphate for the elimination of reactive oxygen species (ROS). While it is now well established that mitochondrial dysfunction plays an important role in the pathophysiology of heart failure, this has been less investigated in atrial myocytes in AF. Considering the high prevalence of AF, investigating the role of mitochondria in this disease may guide the path towards new therapeutic targets. In this review, we discuss the importance of mitochondrial Ca2+ handling in regulating ATP production and mitochondrial ROS emission and how alterations, particularly in these aspects of mitochondrial activity, may play a role in AF. In addition to describing research advances, we highlight areas in which further studies are required to elucidate the role of mitochondria in AF.
Collapse
Affiliation(s)
- Fleur E Mason
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Julius Ryan D Pronto
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Khaled Alhussini
- Department of Thoracic and Cardiovascular Surgery, University Clinic Würzburg, Würzburg, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center Würzburg, University Clinic Würzburg, Am Schwarzenberg 15, 97078, Würzburg, Germany. .,Department of Internal Medicine I, University Clinic Würzburg, Am Schwarzenberg 15, 97078, Würzburg, Germany.
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany. .,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany. .,Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
38
|
Haas Bueno R, Recamonde-Mendoza M. Meta-analysis of Transcriptomic Data Reveals Pathophysiological Modules Involved with Atrial Fibrillation. Mol Diagn Ther 2020; 24:737-751. [PMID: 33095430 DOI: 10.1007/s40291-020-00497-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is a complex disease and affects millions of people around the world. The biological mechanisms that are involved with AF are complex and still need to be fully elucidated. Therefore, we performed a meta-analysis of transcriptome data related to AF to explore these mechanisms aiming at more sensitive and reliable results. METHODS Ten public transcriptomic datasets were downloaded, analyzed for quality control, and individually pre-processed. Differential expression analysis was carried out for each dataset, and the results were meta-analytically aggregated using the rth ordered p value method. We analyzed the final list of differentially expressed genes through network analysis, namely topological and modularity analysis, and functional enrichment analysis. RESULTS The meta-analysis of transcriptomes resulted in 1197 differentially expressed genes, whose protein-protein interaction network presented 39 hubs-bottlenecks and four main identified functional modules. These modules were enriched for 39, 20, 64, and 10 biological pathways involved with the pathophysiology of AF, especially with the disease's structural and electrical remodeling processes. The stress of the endoplasmic reticulum, protein catabolism, oxidative stress, and inflammation are some of the enriched processes. Among hub-bottlenecks genes, which are highly connected and probably have a key role in regulating these processes, HSPA5, ANK2, CTNNB1, and MAPK1 were identified. CONCLUSION Our approach based on transcriptome meta-analysis revealed a set of key genes that demonstrated consistent overall changes in expression patterns associated with AF despite data heterogeneity related, among others, to type of tissue. Further experimental investigation of our findings may shed light on the pathophysiology of the disease and contribute to the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Rodrigo Haas Bueno
- Experimental and Molecular Cardiovascular Laboratory, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil
- Bioinformatics Core, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil
| | - Mariana Recamonde-Mendoza
- Experimental and Molecular Cardiovascular Laboratory, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil.
- Bioinformatics Core, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil.
- Institute of Informatics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| |
Collapse
|
39
|
Podzolkov VI, Tarzimanova AI, Frolova AS. Telocytes and Atrial Fibrillation: From Basic Research to Clinical Practice. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2020. [DOI: 10.20996/1819-6446-2020-08-18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The emergence of new research methods makes it possible to study the tissue, structural, cellular, and molecular causes of atrial fibrillation (AF). Recently, the role of interstitial telocyte cells in the pathogenesis of AF has been actively discussed. Telocytes are a special type of interstitial cells identified in many organs and tissues, including the heart. The roles of telocytes in the myocardium are diverse: they have pacemaker activity, and carry out structural and coordination communication between cells. The ability of these cells to change the speed of the electrical pulse in the atrial and ventricular myocardium has been proven. Telocytes form "atypical" connections with almost all types of cells in the human heart, which collects them in an integrated network. Using electron microscopy, it was found that interstitial cells have different types of connections in the network and can integrate "information" from the vascular and nervous systems, interstitial, immune system, stem cells, progenitor cells, and contractile cardiomyocytes. Currently, the results of studies have been obtained that prove both positive and negative effects of telocytes on the occurrence of various diseases of the cardiovascular system. The role of telocytes in AF arrhythmogenesis remains a subject of discussion. The unique properties of telocytes in providing intercellular contacts, transmitting genetic information, and their ability to regenerate heart tissue are undoubtedly the most promising areas of modern cardiology. There is evidence of both direct and indirect effects of telocytes on the electrophysiological properties of the myocardium. There is no doubt that the development of this area opens up new therapeutic targets for the prevention and treatment of AF.
Collapse
Affiliation(s)
- V. I. Podzolkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - A. I. Tarzimanova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - A. S. Frolova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
40
|
Ciacci A, Falkenberg M, Manani KA, Evans TS, Peters NS, Christensen K. Understanding the transition from paroxysmal to persistent atrial fibrillation. PHYSICAL REVIEW RESEARCH 2020; 2:023311. [PMID: 32607500 PMCID: PMC7326608 DOI: 10.1103/physrevresearch.2.023311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhytmia, characterized by the chaotic motion of electrical wavefronts in the atria. In clinical practice, AF is classified under two primary categories: paroxysmal AF, short intermittent episodes separated by periods of normal electrical activity; and persistent AF, longer uninterrupted episodes of chaotic electrical activity. However, the precise reasons why AF in a given patient is paroxysmal or persistent is poorly understood. Recently, we have introduced the percolation-based Christensen-Manani-Peters (CMP) model of AF which naturally exhibits both paroxysmal and persistent AF, but precisely how these differences emerge in the model is unclear. In this paper, we dissect the CMP model to identify the cause of these different AF classifications. Starting from a mean-field model where we describe AF as a simple birth-death process, we add layers of complexity to the model and show that persistent AF arises from reentrant circuits which exhibit an asymmetry in their probability of activation relative to deactivation. As a result, different simulations generated at identical model parameters can exhibit fibrillatory episodes spanning several orders of magnitude from a few seconds to months. These findings demonstrate that diverse, complex fibrillatory dynamics can emerge from very simple dynamics in models of AF.
Collapse
Affiliation(s)
- Alberto Ciacci
- Blackett Laboratory, Imperial College London, London SW7 2BW, United Kingdom
- Center for Complexity Science, Imperial College London, London SW7 2AZ, United Kingdom
- ElectroCardioMaths Programme, Imperial Centre for Cardiac Engineering, Imperial College London, London W12 0NN, United Kingdom
| | - Max Falkenberg
- Blackett Laboratory, Imperial College London, London SW7 2BW, United Kingdom
- Center for Complexity Science, Imperial College London, London SW7 2AZ, United Kingdom
- ElectroCardioMaths Programme, Imperial Centre for Cardiac Engineering, Imperial College London, London W12 0NN, United Kingdom
| | - Kishan A. Manani
- Blackett Laboratory, Imperial College London, London SW7 2BW, United Kingdom
- Center for Complexity Science, Imperial College London, London SW7 2AZ, United Kingdom
- National Heart and Lung Institute, Imperial College London, London W12 0NN, United Kingdom
| | - Tim S. Evans
- Blackett Laboratory, Imperial College London, London SW7 2BW, United Kingdom
- Center for Complexity Science, Imperial College London, London SW7 2AZ, United Kingdom
| | - Nicholas S. Peters
- ElectroCardioMaths Programme, Imperial Centre for Cardiac Engineering, Imperial College London, London W12 0NN, United Kingdom
| | - Kim Christensen
- Blackett Laboratory, Imperial College London, London SW7 2BW, United Kingdom
- Center for Complexity Science, Imperial College London, London SW7 2AZ, United Kingdom
- ElectroCardioMaths Programme, Imperial Centre for Cardiac Engineering, Imperial College London, London W12 0NN, United Kingdom
| |
Collapse
|
41
|
Increased Neutrophil Granulocyte and Myeloperoxidase Levels Indicate Acute Inflammation Due to the Exposure of Zinc- and Copper-Containing Welding Fumes. J Occup Environ Med 2020; 62:618-627. [PMID: 32404823 DOI: 10.1097/jom.0000000000001905] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Recent studies have shown an increase of C-reactive-protein (CRP) after exposure to zinc- and copper-containing welding fumes. The objective of this study was to determine the effects of exposure to zinc- and copper-containing welding fumes on leukocytes, their subtypes, and myeloperoxidase (MPO). METHODS Serum samples of male volunteers were examined after exposures to welding fumes in two settings: repeated exposure on 4 consecutive days for 6 hours and single exposures for different times (3, 4, 5 hours). RESULTS Neutrophil granulocyte and MPO levels showed increases 24 hours after single and repeated exposures for 6 hours similar to CRP increases reported in literature. Overall leukocyte levels and levels of monocytes and lymphocytes were not significantly affected. CONCLUSIONS This study indicates the involvement of neutrophil granulocytes in welding fume fever additional to mediator related effects.
Collapse
|
42
|
Li LYF, Lou Q, Liu GZ, Lv JC, Yun FX, Li TK, Yang W, Zhao HY, Zhang L, Bai N, Zhan CC, Yu J, Zang YX, Li WM. Sacubitril/valsartan attenuates atrial electrical and structural remodelling in a rabbit model of atrial fibrillation. Eur J Pharmacol 2020; 881:173120. [PMID: 32325147 DOI: 10.1016/j.ejphar.2020.173120] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 04/08/2020] [Accepted: 04/14/2020] [Indexed: 01/09/2023]
Abstract
Atrial structural and electrical remodelling play important roles in atrial fibrillation (AF). Sacubitril/valsartan attenuates cardiac remodelling in heart failure. However, the effect of sacubitril/valsartan on AF is unclear. The aim of this study was to evaluate the effect of sacubitril/valsartan on atrial electrical and structural remodelling in AF and investigate the underlying mechanism of action. Thirty-three rabbits were randomized into sham, RAP, and sac/val groups. HL-1 cells were subjected to control treatment or rapid pacing with or without LBQ657 and valsartan. Echocardiography, atrial electrophysiology, and histological examination were performed. The concentration of Ca2+ and expression levels of calcineurin, NFAT, p-NFAT, Cav1.2, collagen Ⅰ and Ⅲ, ANP, BNP, CNP, NT-proBNP, and ST2 in HL-1 cells, and IcaL in left atrial cells, were determined. We observed that compared to that in the sham group, the atrium and right ventricle were enlarged, myocardial fibrosis was markedly higher, AF inducibility was significantly elevated, and atrial effective refractory periods were shortened in the RAP group. These effects were significantly reversed by sacubitril/valsartan. Compared to that in the sham group, collagen Ⅰ and Ⅲ, NT-proBNP, ST2, calcineurin, and NFAT were significantly up-regulated, while p-NFAT and Cav1.2 were down-regulated in the RAP group, and sacubitril/valsartan inhibited these changes. Ca2+ concentration increased and ICaL density decreased in in vivo and in vitro AF models, reversed by sacubitril/valsartan. Sacubitril/valsartan attenuates atrial electrical remodelling and ameliorates structure remodelling in AF. This study paves the way for the possibility of clinical use of sacubitril/valsartan in AF patients.
Collapse
Affiliation(s)
- Lu-Yi-Fei Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Qi Lou
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Guang-Zhong Liu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jia-Chen Lv
- Department of Colorectal Surgery, The Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Feng-Xiang Yun
- Department of Internal Critic Care, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Tian-Kai Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wen Yang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Hong-Yan Zhao
- Department of Cardiology, The People's Hospital of Liaoning Province, Shenyang, China
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Nan Bai
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Cheng-Chuang Zhan
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jia Yu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yan-Xiang Zang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wei-Min Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
43
|
Linking cellular energy state to atrial fibrillation pathogenesis: Potential role of adenosine monophosphate-activated protein kinase. Heart Rhythm 2020; 17:1398-1404. [PMID: 32268208 DOI: 10.1016/j.hrthm.2020.03.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 03/28/2020] [Indexed: 01/01/2023]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is the cellular stress-sensing molecule. Apart from maintaining cellular energy balance, AMPK controls expression and regulation of ion channels and ion transporters, including cytosolic Ca2+ handling proteins. Emerging evidence suggests that metabolic impairment plays a crucial role in the pathogenesis of atrial fibrillation. AMPK activation is thought to be protective by preventing metabolic stress, favorably modulating membrane electrophysiology including cytosolic Ca2+ dynamics; preventing cellular growth; and hypertrophic remodeling. This review considers current concepts and evidence from clinical and experimental studies regarding the role of AMPK in atrial fibrillation.
Collapse
|
44
|
Coppini R, Santini L, Palandri C, Sartiani L, Cerbai E, Raimondi L. Pharmacological Inhibition of Serine Proteases to Reduce Cardiac Inflammation and Fibrosis in Atrial Fibrillation. Front Pharmacol 2019; 10:1420. [PMID: 31956307 PMCID: PMC6951407 DOI: 10.3389/fphar.2019.01420] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/07/2019] [Indexed: 12/18/2022] Open
Abstract
Systemic inflammation correlates with an increased risk of atrial fibrillation (AF) and thrombogenesis. Systemic inflammation alters vessel permeability, allowing inflammatory and immune cell migration toward target organs, including the heart. Among inflammatory cells infiltrating the atria, macrophages and mast cell have recently attracted the interest of basic researchers due to the pathogenic mechanisms triggered by their activation. This chemotactic invasion is likely implicated in short- and long-term changes in cardiac cell-to-cell communication and in triggering fibrous tissue accumulation in the atrial myocardium and electrophysiological re-arrangements of atrial cardiomyocytes, thus favoring the onset and progression of AF. Serine proteases are a large and heterogeneous class of proteases involved in several processes that are important for cardiac function and are involved in cardiac diseases, such as (i) coagulation, (ii) fibrinolysis, (iii) extracellular matrix degradation, (iv) activation of receptors (i.e., protease-activated receptors [PPARs]), and (v) modulation of the activity of endogenous signals. The recognition of serine proteases substrates and their involvement in inflammatory/profibrotic mechanisms allowed the identification of novel cardio-protective mechanisms for commonly used drugs that inhibit serine proteases. The aim of this review is to summarize knowledge on the role of inflammation and fibrosis as determinants of AF. Moreover, we will recapitulate current findings on the role of serine proteases in the pathogenesis of AF and the possible beneficial effects of drugs inhibiting serine proteases in reducing the risk of AF through decrease of cardiac inflammation and fibrosis. These drugs include thrombin and factor Xa inhibitors (used as oral anticoagulants), dipeptidyl-peptidase 4 (DPP4) inhibitors, used for type-2 diabetes, as well as novel experimental inhibitors of mast cell chymases.
Collapse
Affiliation(s)
- Raffaele Coppini
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| | - Lorenzo Santini
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| | - Chiara Palandri
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| | - Laura Sartiani
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| | - Elisabetta Cerbai
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| | - Laura Raimondi
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| |
Collapse
|
45
|
Khidirova LD, Yakhontov DA, Zenin SA, Kuropii TS. The impact of chronic obstructive pulmonary disease and hypertension on the development and progression of atrial fibrillation. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2019. [DOI: 10.15829/1728-8800-2019-5-138-144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Affiliation(s)
| | | | - S. A. Zenin
- Novosibirsk Regional Cardiology Clinical Dispensary
| | | |
Collapse
|
46
|
Holzwirth E, Kornej J, Erbs S, Obradovic D, Bollmann A, Hindricks G, Thiele H, Büttner P. Myeloperoxidase in atrial fibrillation: association with progression, origin and influence of renin-angiotensin system antagonists. Clin Res Cardiol 2019; 109:324-330. [PMID: 31236695 DOI: 10.1007/s00392-019-01512-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/17/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND Myeloperoxidase (MPO), secreted by neutrophils under inflammatory conditions, is elevated in atrial fibrillation (AF). MPO may be involved in atrial remodeling that underpins AF progression characterized by a switch from paroxysmal to persistent AF and the formation of low-voltage areas (LVA). MPO levels are modulated by renin-angiotensin system antagonists (RAS-A), commonly used to treat AF comorbidities, and are associated with reduced AF incidence, implicating a potential link. OBJECTIVE We investigated MPO levels in progressing AF in peripheral and left atrial (LA) blood and analyzed a potential effect of RAS-A. METHODS Samples of AF patients were collected from the femoral vein and the LA during catheter ablation (n = 121) and at follow-up (n = 23). No-AF probands (n = 37) served as controls. MPO was determined using commercial ELISA. RESULTS MPO levels were significantly increased in AF patients compared to controls (median, 27.7 ng/ml (IQR 14.3-66.6) versus 12.6 (IQR 9.9-17.7), p < 0.001), without differences between clinical AF progression phenotypes. MPO concentration was tenfold higher in LA than periphery (279.2 ng/ml (IQR 202.2-342.9) versus 27.7 ng/ml (IQR 14.3-65.9), p < 0.001). MPO remained increased at midterm follow-up irrespective of rhythm outcome. RAS-A was associated with significantly lower peripheral (22.2 ng/ml (IQR 12.7-48.2) versus 37.1 ng/ml (IQR 18.2-85.2), p < 0.05) MPO levels in AF patients. CONCLUSION The pro-fibrotic enzyme MPO is generally elevated in AF patients irrespective of AF type, the presence of LVA or midterm rhythm outcome. Our data suggest that MPO may directly originate from the LA. RAS-A decrease peripheral MPO levels in AF patients.
Collapse
Affiliation(s)
- Erik Holzwirth
- Department of Cardiology, Heart Center Leipzig at University Leipzig, Strümpellstraße 39, 04289, Leipzig, Germany
| | - Jelena Kornej
- Department of Electrophysiology, Heart Center Leipzig at University Leipzig, Leipzig, Germany
| | - Sandra Erbs
- Department of Cardiology, Heart Center Leipzig at University Leipzig, Strümpellstraße 39, 04289, Leipzig, Germany
| | - Danilo Obradovic
- Department of Cardiology, Heart Center Leipzig at University Leipzig, Strümpellstraße 39, 04289, Leipzig, Germany
| | - Andreas Bollmann
- Department of Electrophysiology, Heart Center Leipzig at University Leipzig, Leipzig, Germany
| | - Gerhard Hindricks
- Department of Electrophysiology, Heart Center Leipzig at University Leipzig, Leipzig, Germany
| | - Holger Thiele
- Department of Cardiology, Heart Center Leipzig at University Leipzig, Strümpellstraße 39, 04289, Leipzig, Germany
| | - Petra Büttner
- Department of Cardiology, Heart Center Leipzig at University Leipzig, Strümpellstraße 39, 04289, Leipzig, Germany.
| |
Collapse
|
47
|
Ndrepepa G. Myeloperoxidase - A bridge linking inflammation and oxidative stress with cardiovascular disease. Clin Chim Acta 2019; 493:36-51. [PMID: 30797769 DOI: 10.1016/j.cca.2019.02.022] [Citation(s) in RCA: 253] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/20/2019] [Accepted: 02/20/2019] [Indexed: 12/17/2022]
Abstract
Myeloperoxidase (MPO) is a member of the superfamily of heme peroxidases that is mainly expressed in neutrophils and monocytes. MPO-derived reactive species play a key role in neutrophil antimicrobial activity and human defense against various pathogens primarily by participating in phagocytosis. Elevated MPO levels in circulation are associated with inflammation and increased oxidative stress. Multiple lines of evidence suggest an association between MPO and cardiovascular disease (CVD) including coronary artery disease, congestive heart failure, arterial hypertension, pulmonary arterial hypertension, peripheral arterial disease, myocardial ischemia/reperfusion-related injury, stroke, cardiac arrhythmia and venous thrombosis. Elevated MPO levels are associated with a poor prognosis including increased risk for overall and CVD-related mortality. Elevated MPO may signify an increased risk for CVD for at least 2 reasons. First, low-grade inflammation and increased oxidative stress coexist with many metabolic abnormalities and comorbidities and consequently an elevated MPO level may represent an increased cardiometabolic risk in general. Second, MPO produces a large number of highly reactive species which can attack, destroy or modify the function of every known cellular component. The most common MPO actions relevant to CVD are generation of dysfunctional lipoproteins with an increased atherogenicity potential, reduced NO availability, endothelial dysfunction, impaired vasoreactivity and atherosclerotic plaque instability. These actions strongly suggest that MPO is directly involved in the pathophysiology of CVD. In this regard MPO may be seen as a mediator or an instrument through which inflammation promotes CVD at molecular and cellular level. Clinical value of MPO therapeutic inhibition remains to be tested.
Collapse
Affiliation(s)
- Gjin Ndrepepa
- Department of Adult Cardiology, Deutsches Herzzentrum München, Technische Universität, Lazarettstrasse 36, 80636 Munich, Germany.
| |
Collapse
|
48
|
Li G, Yang Q, Yang Y, Yang G, Wan J, Ma Z, Du L, Sun Y, Ζhang G. Laminar shear stress alters endothelial KCa2.3 expression in H9c2 cells partially via regulating the PI3K/Akt/p300 axis. Int J Mol Med 2019; 43:1289-1298. [PMID: 30664154 PMCID: PMC6365081 DOI: 10.3892/ijmm.2019.4063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 01/10/2019] [Indexed: 12/11/2022] Open
Abstract
In cardiac tissues, myoblast atrial myocytes continue to be exposed to mechanical forces including shear stress. However, little is known about the effects of shear stress on atrial myocytes, particularly on ion channel function, in association with disease. The present study demonstrated that the Ca2+-activated K+ channel (KCa)2.3 serves a vital role in regulating arterial tone. As increased intracellular Ca2+ levels and activation of histone acetyltransferase p300 (p300) are early responses to laminar shear stress (LSS) that result in the transcriptional activation of genes, the role of p300 and the phosphoinositide3-kinase (PI3K)/protein kinase B (Akt) pathway, an intracellular pathway that promotes the growth and proliferation rather than the differentiation of adult cells, in the LSS-dependent regulation of KCa2.3 in cardiac myoblasts was examined. In cultured H9c2 cells, exposure to LSS (15 dyn/cm2) for 12 h markedly increased KCa2.3 mRNA expression. Inhibiting PI3K attenuated the LSS-induced increases in the expression and channel activity of KCa2.3, and decreased the phosphorylation levels of p300. The upregulation of these channels was abolished by the inhibition of Akt through decreasing p300 phosphorylation. ChIP assays indicated that p300 was recruited to the promoter region of the KCa2.3 gene. Therefore, the PI3K/Akt/p300 axis serves a crucial role in the LSS-dependent induction of KCa2.3 expression, by regulating cardiac myoblast function and adaptation to hemodynamic changes. The key novel insights gained from the present study are: i) KCa2.3 was upregulated in patients with atrial fibrillation (AF) and in patients with AF combined with mitral value disease; ii) LSS induced a profound upregulation of KCa2.3 mRNA and protein expression in H9c2 cells; iii) PI3K activation was associated with LSS-induced upregulation of the KCa2.3 channel; iv) PI3K activation was mediated by PI3K/Akt-dependent Akt activation; and v) LSS induction of KCa2.3 involved the binding of p300 to transcription factors in the promoter region of the KCa2.3 gene.
Collapse
Affiliation(s)
- Guojian Li
- Department of Vascular Surgery, The Second People's Hospital of Yunnan Province, Kunming Medical University, Kunming, Yunnan 650200, P.R. China
| | - Qionghui Yang
- Department of Pharmacy, The Third People's Hospital of Yunnan Province, Kunming, Yunnan 650200, P.R. China
| | - Yong Yang
- Department of Vascular Surgery, The Second People's Hospital of Yunnan Province, Kunming Medical University, Kunming, Yunnan 650200, P.R. China
| | - Guokai Yang
- Department of Vascular Surgery, The Second People's Hospital of Yunnan Province, Kunming Medical University, Kunming, Yunnan 650200, P.R. China
| | - Jia Wan
- Department of Vascular Surgery, The Second People's Hospital of Yunnan Province, Kunming Medical University, Kunming, Yunnan 650200, P.R. China
| | - Zhenhuan Ma
- Department of Vascular Surgery, The Second People's Hospital of Yunnan Province, Kunming Medical University, Kunming, Yunnan 650200, P.R. China
| | - Lingjuan Du
- Department of Vascular Surgery, The Second People's Hospital of Yunnan Province, Kunming Medical University, Kunming, Yunnan 650200, P.R. China
| | - Yi Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Guimin Ζhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| |
Collapse
|
49
|
Meng L, Wong R, Tsui MY, Tse G, Li G, Liu T, Lip GYH. Urinary Biomarkers of Oxidative Stress in Atrial Fibrillation. THE OPEN BIOMARKERS JOURNAL 2018; 8:24-33. [DOI: 10.2174/1875318301808010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/08/2018] [Accepted: 11/12/2018] [Indexed: 10/11/2023]
Abstract
There is increasing evidence from molecular studies to support the role of inflammation and increased oxidative stress that produce structural and electrical atrial remodeling to produce Atrial Fbrillation (AF). Oxidative damage to cardiomyocytes yields chemical substances that are secreted in urine. These substances can serve as biomarkers that can be measured, potentially allowing clinicians to quantify oxidative damage to the heart.
Collapse
|
50
|
Kornej J, Büttner P, Hammer E, Engelmann B, Dinov B, Sommer P, Husser D, Hindricks G, Völker U, Bollmann A. Circulating proteomic patterns in AF related left atrial remodeling indicate involvement of coagulation and complement cascade. PLoS One 2018; 13:e0198461. [PMID: 30496173 PMCID: PMC6264811 DOI: 10.1371/journal.pone.0198461] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 11/12/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Left atrial (LA) electro-anatomical remodeling and diameter increase in atrial fibrillation (AF) indicates disease progression and is associated with poor therapeutic success. Furthermore, AF leads to a hypercoagulable state, which in turn promotes the development of a substrate for AF and disease progression in the experimental setting. The aim of this study was to identify pathways associated with LA remodeling in AF patients using untargeted proteomics approach. METHODS Peripheral blood samples of 48 patients (62±10 years, 63% males, 59% persistent AF) undergoing AF catheter ablation were collected before ablation. 23 patients with left atrial low voltage areas (LVA), defined as <0.5 mV, and 25 patients without LVA were matched for age, gender and CHA2DS2-VASc score. Untargeted proteome analysis was performed using LC-ESI-Tandem mass spectrometry in a label free intensity based workflow. Significantly different abundant proteins were identified and used for pathway analysis and protein-protein interaction analysis. RESULTS Analysis covered 280 non-redundant circulating plasma proteins. The presence of LVA correlated with 30 differentially abundant proteins of coagulation and complement cascade (q<0.05). CONCLUSIONS This pilot proteomic study identified plasma protein candidates associated with electro-anatomical remodeling in AF and pointed towards an imbalance in coagulation and complement pathway, tissue remodeling and inflammation.
Collapse
Affiliation(s)
- Jelena Kornej
- Department of Electrophysiology, Heart Center Leipzig, University of Leipzig, Leipzig, Germany
- * E-mail:
| | - Petra Büttner
- Department of Electrophysiology, Heart Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Elke Hammer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Beatrice Engelmann
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Borislav Dinov
- Department of Electrophysiology, Heart Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Philipp Sommer
- Department of Electrophysiology, Heart Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Daniela Husser
- Department of Electrophysiology, Heart Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Gerhard Hindricks
- Department of Electrophysiology, Heart Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Andreas Bollmann
- Department of Electrophysiology, Heart Center Leipzig, University of Leipzig, Leipzig, Germany
| |
Collapse
|