1
|
Miotto MC, Reiken S, Wronska A, Yuan Q, Dridi H, Liu Y, Weninger G, Tchagou C, Marks AR. Structural basis for ryanodine receptor type 2 leak in heart failure and arrhythmogenic disorders. Nat Commun 2024; 15:8080. [PMID: 39278969 PMCID: PMC11402997 DOI: 10.1038/s41467-024-51791-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 08/12/2024] [Indexed: 09/18/2024] Open
Abstract
Heart failure, the leading cause of mortality and morbidity in the developed world, is characterized by cardiac ryanodine receptor 2 channels that are hyperphosphorylated, oxidized, and depleted of the stabilizing subunit calstabin-2. This results in a diastolic sarcoplasmic reticulum Ca2+ leak that impairs cardiac contractility and triggers arrhythmias. Genetic mutations in ryanodine receptor 2 can also cause Ca2+ leak, leading to arrhythmias and sudden cardiac death. Here, we solved the cryogenic electron microscopy structures of ryanodine receptor 2 variants linked either to heart failure or inherited sudden cardiac death. All are in the primed state, part way between closed and open. Binding of Rycal drugs to ryanodine receptor 2 channels reverts the primed state back towards the closed state, decreasing Ca2+ leak, improving cardiac function, and preventing arrhythmias. We propose a structural-physiological mechanism whereby the ryanodine receptor 2 channel primed state underlies the arrhythmias in heart failure and arrhythmogenic disorders.
Collapse
Affiliation(s)
- Marco C Miotto
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA.
| | - Steven Reiken
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| | - Anetta Wronska
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| | - Qi Yuan
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| | - Haikel Dridi
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| | - Yang Liu
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| | - Gunnar Weninger
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| | - Carl Tchagou
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| | - Andrew R Marks
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA.
| |
Collapse
|
2
|
Mo W, Donahue JK. Gene therapy for atrial fibrillation. J Mol Cell Cardiol 2024; 196:84-93. [PMID: 39270930 DOI: 10.1016/j.yjmcc.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 08/19/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia in adults. Current limitations of pharmacological and ablative therapies motivate the development of novel therapies as next generation treatments for AF. The arrhythmia mechanisms creating and sustaining AF are key elements in the development of this novel treatment. Gene therapy provides a useful platform that allows us to regulate the mechanisms of interest using a suitable transgene(s), vector, and delivery method. Effective gene therapy strategies in the literature have targeted maladaptive electrical or structural remodeling that increase vulnerability to AF. In this review, we will summarize key elements of gene therapy for AF, including molecular targets, gene transfer vectors, atrial gene delivery and preclinical efficacy and toxicity testing. Recent advances and challenges in the field will be also discussed.
Collapse
Affiliation(s)
- Weilan Mo
- From the Division of Cardiology, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - J Kevin Donahue
- From the Division of Cardiology, University of Massachusetts Medical School, Worcester, MA, United States of America.
| |
Collapse
|
3
|
Li TL, Zhu NN, Yin Z, Sun J, Guo JP, Yuan HT, Shi XM, Guo HY, Li SX, Shan ZL. Transcriptomic analysis of epicardial adipose tissue reveals the potential crosstalk genes and immune relationship between type 2 diabetes mellitus and atrial fibrillation. Gene 2024; 920:148528. [PMID: 38703871 DOI: 10.1016/j.gene.2024.148528] [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: 12/02/2023] [Revised: 03/27/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND The complex relationship between atrial fibrillation (AF) and type 2 diabetes mellitus (T2DM) suggests a potential role for epicardial adipose tissue (EAT) that requires further investigation. This study employs bioinformatics and experimental approaches to clarify EAT's role in linking T2DM and AF, aiming to unravel the biological mechanisms involved. METHOD Bioinformatics analysis initially identified common differentially expressed genes (DEGs) in EAT from T2DM and AF datasets. Pathway enrichment and network analyses were then performed to determine the biological significance and network connections of these DEGs. Hub genes were identified through six CytoHubba algorithms and subsequently validated biologically, with further in-depth analyses confirming their roles and interactions. Experimentally, db/db mice were utilized to establish a T2DM model. AF induction was executed via programmed transesophageal electrical stimulation and burst pacing, focusing on comparing the incidence and duration of AF. Frozen sections and Hematoxylin and Eosin (H&E) staining illuminated the structures of the heart and EAT. Moreover, quantitative PCR (qPCR) measured the expression of hub genes. RESULTS The study identified 106 DEGs in EAT from T2DM and AF datasets, underscoring significant pathways in energy metabolism and immune regulation. Three hub genes, CEBPZ, PAK1IP1, and BCCIP, emerged as pivotal in this context. In db/db mice, a marked predisposition towards AF induction and extended duration was observed, with HE staining verifying the presence of EAT. Additionally, qPCR validated significant changes in hub genes expression in db/db mice EAT. In-depth analysis identified 299 miRNAs and 33 TFs as potential regulators, notably GRHL1 and MYC. GeneMANIA analysis highlighted the hub genes' critical roles in stress responses and leukocyte differentiation, while immune profile correlations highlighted their impact on mast cells and neutrophils, emphasizing the genes' significant influence on immune regulation within the context of T2DM and AF. CONCLUSION This investigation reveals the molecular links between T2DM and AF with a focus on EAT. Targeting these pathways, especially EAT-related ones, may enable personalized treatments and improved outcomes.
Collapse
Affiliation(s)
- Tian-Lun Li
- Postgraduate School, Medical School of Chinese PLA, Beijing, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Na-Na Zhu
- Postgraduate School, Medical School of Chinese PLA, Beijing, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhao Yin
- Postgraduate School, Medical School of Chinese PLA, Beijing, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jiao Sun
- Postgraduate School, Medical School of Chinese PLA, Beijing, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jian-Pin Guo
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hong-Tao Yuan
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiang-Min Shi
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hong-Yang Guo
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shi-Xing Li
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhao-Liang Shan
- Postgraduate School, Medical School of Chinese PLA, Beijing, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
4
|
Schönmehl R, Mendelsohn DH, Winter L, Pabel S, Niedermair T, Evert K, Cheung WH, Wong RMY, Schmitt VH, Keller K, Barsch F, Dietl A, Gummert JF, Schramm R, Sossalla S, Brochhausen C. Comparative Analysis of Mitochondria Surrounding the Intercalated Discs in Heart Diseases-An Ultrastructural Pilot Study. Int J Mol Sci 2024; 25:7644. [PMID: 39062885 PMCID: PMC11277158 DOI: 10.3390/ijms25147644] [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: 02/20/2024] [Revised: 06/28/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Mitochondria play a crucial role in adapting to fluctuating energy demands, particularly in various heart diseases. This study investigates mitochondrial morphology near intercalated discs in left ventricular (LV) heart tissues, comparing samples from patients with sinus rhythm (SR), atrial fibrillation (AF), dilated cardiomyopathy (DCM), and ischemic cardiomyopathy (ICM). METHODS Transmission electron microscopy was used to analyze mitochondria within 0-3.5 μm and 3.5-7 μm of intercalated discs in 9 SR, 10 AF, 9 DCM, and 8 ICM patient samples. Parameters included mean size in µm2 and elongation, count, percental mitochondrial area in the measuring frame, and a conglomeration score. RESULTS AF patients exhibited higher counts of small mitochondria in the LV myocardium, resembling SR. DCM and ICM groups had fewer, larger, and often hydropic mitochondria. Accumulation rates and percental mitochondrial area were similar across groups. Significant positive correlations existed between other defects/size and hydropic mitochondria and between count/area and conglomeration score, while negative correlations between count and size/other defects and between hydropic mitochondria and count could be seen as well. CONCLUSION Mitochondrial parameters in the LV myocardium of AF patients were similar to those of SR patients, while DCM and ICM displayed distinct changes, including a decrease in number, an increase in size, and compromised mitochondrial morphology. Further research is needed to fully elucidate the pathophysiological role of mitochondrial morphology in different heart diseases, providing deeper insights into potential therapeutic targets and interventions.
Collapse
Affiliation(s)
- Rebecca Schönmehl
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Daniel H. Mendelsohn
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Lina Winter
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Steffen Pabel
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Tanja Niedermair
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany (K.E.)
- Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
| | - Katja Evert
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany (K.E.)
| | - Wing-Hoi Cheung
- Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ronald Man Yeung Wong
- Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Volker H. Schmitt
- Department of Cardiology, University Medical Centre, Johannes Gutenberg University of Mainz, 55131 Mainz, Germany (K.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, 55131 Mainz, Germany
| | - Karsten Keller
- Department of Cardiology, University Medical Centre, Johannes Gutenberg University of Mainz, 55131 Mainz, Germany (K.K.)
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Department of Sports Medicine, Medical Clinic VII, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Friedrich Barsch
- Medical Center, Faculty of Medicine, Institute for Exercise and Occupational Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Alexander Dietl
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Jan F. Gummert
- Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center North Rhine-Westphalia, Ruhr University Bochum, 32545 Bad Oeynhausen, Germany
| | - René Schramm
- Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center North Rhine-Westphalia, Ruhr University Bochum, 32545 Bad Oeynhausen, Germany
| | - Samuel Sossalla
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
- Departments of Cardiology at Kerckhoff Heart and Lung Center, Bad Nauheim and University of Giessen, 61231 Bad Nauheim, Germany
| | - Christoph Brochhausen
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
5
|
Murphy MB, Yang Z, Subati T, Farber-Eger E, Kim K, Blackwell DJ, Fleming MR, Stark JM, Van Amburg JC, Woodall KK, Van Beusecum JP, Agrawal V, Smart CD, Pitzer A, Atkinson JB, Fogo AB, Bastarache JA, Kirabo A, Wells QS, Madhur MS, Barnett JV, Murray KT. LNK/SH2B3 loss of function increases susceptibility to murine and human atrial fibrillation. Cardiovasc Res 2024; 120:899-913. [PMID: 38377486 PMCID: PMC11218690 DOI: 10.1093/cvr/cvae036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/31/2023] [Accepted: 10/07/2023] [Indexed: 02/22/2024] Open
Abstract
AIMS The lymphocyte adaptor protein (LNK) is a negative regulator of cytokine and growth factor signalling. The rs3184504 variant in SH2B3 reduces LNK function and is linked to cardiovascular, inflammatory, and haematologic disorders, including stroke. In mice, deletion of Lnk causes inflammation and oxidative stress. We hypothesized that Lnk-/- mice are susceptible to atrial fibrillation (AF) and that rs3184504 is associated with AF and AF-related stroke in humans. During inflammation, reactive lipid dicarbonyls are the major components of oxidative injury, and we further hypothesized that these mediators are critical drivers of the AF substrate in Lnk-/- mice. METHODS AND RESULTS Lnk-/- or wild-type (WT) mice were treated with vehicle or 2-hydroxybenzylamine (2-HOBA), a dicarbonyl scavenger, for 3 months. Compared with WT, Lnk-/- mice displayed increased AF duration that was prevented by 2-HOBA. In the Lnk-/- atria, action potentials were prolonged with reduced transient outward K+ current, increased late Na+ current, and reduced peak Na+ current, pro-arrhythmic effects that were inhibited by 2-HOBA. Mitochondrial dysfunction, especially for Complex I, was evident in Lnk-/- atria, while scavenging lipid dicarbonyls prevented this abnormality. Tumour necrosis factor-α (TNF-α) and interleukin-1 beta (IL-1β) were elevated in Lnk-/- plasma and atrial tissue, respectively, both of which caused electrical and bioenergetic remodelling in vitro. Inhibition of soluble TNF-α prevented electrical remodelling and AF susceptibility, while IL-1β inhibition improved mitochondrial respiration but had no effect on AF susceptibility. In a large database of genotyped patients, rs3184504 was associated with AF, as well as AF-related stroke. CONCLUSION These findings identify a novel role for LNK in the pathophysiology of AF in both experimental mice and humans. Moreover, reactive lipid dicarbonyls are critical to the inflammatory AF substrate in Lnk-/- mice and mediate the pro-arrhythmic effects of pro-inflammatory cytokines, primarily through electrical remodelling.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Action Potentials/drug effects
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Atrial Fibrillation/metabolism
- Atrial Fibrillation/physiopathology
- Atrial Fibrillation/genetics
- Benzylamines/pharmacology
- Disease Models, Animal
- Genetic Predisposition to Disease
- Heart Rate/drug effects
- Inflammation Mediators/metabolism
- Interleukin-1beta/metabolism
- Interleukin-1beta/genetics
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Mitochondria, Heart/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Oxidative Stress/drug effects
- Phenotype
- Signal Transduction
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/genetics
Collapse
Affiliation(s)
- Matthew B Murphy
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Zhenjiang Yang
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Tuerdi Subati
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Eric Farber-Eger
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
| | - Kyungsoo Kim
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Daniel J Blackwell
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Matthew R Fleming
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
| | - Joshua M Stark
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Joseph C Van Amburg
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Kaylen K Woodall
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Justin P Van Beusecum
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Vineet Agrawal
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
| | - Charles D Smart
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Ashley Pitzer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - James B Atkinson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, 1161 21 Avenue South, Nashville, TN 37232, USA
| | - Agnes B Fogo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, 1161 21 Avenue South, Nashville, TN 37232, USA
| | - Julie A Bastarache
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Quinn S Wells
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, 2525 West End Avenue, Nashville, TN 37203, USA
| | - Meena S Madhur
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Joey V Barnett
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Katherine T Murray
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| |
Collapse
|
6
|
Ayagama T, Charles PD, Bose SJ, Boland B, Priestman DA, Aston D, Berridge G, Fischer R, Cribbs AP, Song Q, Mirams GR, Amponsah K, Heather L, Galione A, Herring N, Kramer H, Capel RA, Platt FM, Schotten U, Verheule S, Burton RA. Compartmentalization proteomics revealed endolysosomal protein network changes in a goat model of atrial fibrillation. iScience 2024; 27:109609. [PMID: 38827406 PMCID: PMC11141153 DOI: 10.1016/j.isci.2024.109609] [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: 08/30/2023] [Revised: 02/07/2024] [Accepted: 03/25/2024] [Indexed: 06/04/2024] Open
Abstract
Endolysosomes (EL) are known for their role in regulating both intracellular trafficking and proteostasis. EL facilitate the elimination of damaged membranes, protein aggregates, membranous organelles and play an important role in calcium signaling. The specific role of EL in cardiac atrial fibrillation (AF) is not well understood. We isolated atrial EL organelles from AF goat biopsies and conducted a comprehensive integrated omics analysis to study the EL-specific proteins and pathways. We also performed electron tomography, protein and enzyme assays on these biopsies. Our results revealed the upregulation of the AMPK pathway and the expression of EL-specific proteins that were not found in whole tissue lysates, including GAA, DYNLRB1, CLTB, SIRT3, CCT2, and muscle-specific HSPB2. We also observed structural anomalies, such as autophagic-vacuole formation, irregularly shaped mitochondria, and glycogen deposition. Our results provide molecular information suggesting EL play a role in AF disease process over extended time frames.
Collapse
Affiliation(s)
- Thamali Ayagama
- Department of Pharmacology, University of Oxford, Oxford, UK
| | | | - Samuel J. Bose
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Barry Boland
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | | | - Daniel Aston
- Department of Anaesthesia and Critical Care, Royal Papworth Hospital NHS Foundation Trust, Papworth Road, Cambridge CB2 0AY, UK
| | | | - Roman Fischer
- Target Discovery Institute, University of Oxford, Oxford, UK
| | - Adam P. Cribbs
- Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Headington OX3 7LD, UK
| | - Qianqian Song
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Gary R. Mirams
- Centre for Mathematical Medicine & Biology, Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Kwabena Amponsah
- Centre for Mathematical Medicine & Biology, Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Lisa Heather
- Department of Physiology, Anatomy and Genetics, , University of Oxford, South Park Road, Oxford OX1 3PT, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Neil Herring
- Department of Physiology, Anatomy and Genetics, , University of Oxford, South Park Road, Oxford OX1 3PT, UK
| | - Holger Kramer
- Mass spectrometry Facility, The MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | | | | | - Ulrich Schotten
- Departments of Physiology and Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Sander Verheule
- Departments of Physiology and Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Rebecca A.B. Burton
- Department of Pharmacology, University of Oxford, Oxford, UK
- University of Liverpool, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, Liverpool, UK
| |
Collapse
|
7
|
Sarkar A, Fanous KI, Marei I, Ding H, Ladjimi M, MacDonald R, Hollenberg MD, Anderson TJ, Hill MA, Triggle CR. Repurposing Metformin for the Treatment of Atrial Fibrillation: Current Insights. Vasc Health Risk Manag 2024; 20:255-288. [PMID: 38919471 PMCID: PMC11198029 DOI: 10.2147/vhrm.s391808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
Metformin is an orally effective anti-hyperglycemic drug that despite being introduced over 60 years ago is still utilized by an estimated 120 to 150 million people worldwide for the treatment of type 2 diabetes (T2D). Metformin is used off-label for the treatment of polycystic ovary syndrome (PCOS) and for pre-diabetes and weight loss. Metformin is a safe, inexpensive drug with side effects mostly limited to gastrointestinal issues. Prospective clinical data from the United Kingdom Prospective Diabetes Study (UKPDS), completed in 1998, demonstrated that metformin not only has excellent therapeutic efficacy as an anti-diabetes drug but also that good glycemic control reduced the risk of micro- and macro-vascular complications, especially in obese patients and thereby reduced the risk of diabetes-associated cardiovascular disease (CVD). Based on a long history of clinical use and an excellent safety record metformin has been investigated to be repurposed for numerous other diseases including as an anti-aging agent, Alzheimer's disease and other dementias, cancer, COVID-19 and also atrial fibrillation (AF). AF is the most frequently diagnosed cardiac arrythmia and its prevalence is increasing globally as the population ages. The argument for repurposing metformin for AF is based on a combination of retrospective clinical data and in vivo and in vitro pre-clinical laboratory studies. In this review, we critically evaluate the evidence that metformin has cardioprotective actions and assess whether the clinical and pre-clinical evidence support the use of metformin to reduce the risk and treat AF.
Collapse
Affiliation(s)
- Aparajita Sarkar
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Kareem Imad Fanous
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Isra Marei
- Department of Pharmacology & Medical Education, Weill Cornell Medicine- Qatar, Doha, Qatar
| | - Hong Ding
- Department of Pharmacology & Medical Education, Weill Cornell Medicine- Qatar, Doha, Qatar
| | - Moncef Ladjimi
- Department of Biochemistry & Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Ross MacDonald
- Health Sciences Library, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Morley D Hollenberg
- Department of Physiology & Pharmacology, and Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Todd J Anderson
- Department of Cardiac Sciences and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michael A Hill
- Dalton Cardiovascular Research Center & Department of Medical Pharmacology & Physiology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Chris R Triggle
- Department of Pharmacology & Medical Education, Weill Cornell Medicine- Qatar, Doha, Qatar
| |
Collapse
|
8
|
Zhong J, Chen H, Liu Q, Zhou S, Liu Z, Xiao Y. GLP-1 receptor agonists and myocardial metabolism in atrial fibrillation. J Pharm Anal 2024; 14:100917. [PMID: 38799233 PMCID: PMC11127228 DOI: 10.1016/j.jpha.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 10/15/2023] [Accepted: 12/07/2023] [Indexed: 05/29/2024] Open
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia. Many medical conditions, including hypertension, diabetes, obesity, sleep apnea, and heart failure (HF), increase the risk for AF. Cardiomyocytes have unique metabolic characteristics to maintain adenosine triphosphate production. Significant changes occur in myocardial metabolism in AF. Glucagon-like peptide-1 receptor agonists (GLP-1RAs) have been used to control blood glucose fluctuations and weight in the treatment of type 2 diabetes mellitus (T2DM) and obesity. GLP-1RAs have also been shown to reduce oxidative stress, inflammation, autonomic nervous system modulation, and mitochondrial function. This article reviews the changes in metabolic characteristics in cardiomyocytes in AF. Although the clinical trial outcomes are unsatisfactory, the findings demonstrate that GLP-1 RAs can improve myocardial metabolism in the presence of various risk factors, lowering the incidence of AF.
Collapse
Affiliation(s)
- Jiani Zhong
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Xiangya School of Medicine, Central South University, Changsha, 410008, China
| | - Hang Chen
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Xiangya School of Medicine, Central South University, Changsha, 410008, China
| | - Qiming Liu
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Shenghua Zhou
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Zhenguo Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, 65211, USA
| | - Yichao Xiao
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| |
Collapse
|
9
|
Cao Y, Cui L, Tuo S, Liu H, Cui S. Resveratrol mediates mitochondrial function through the sirtuin 3 pathway to improve abnormal metabolic remodeling in atrial fibrillation. Eur J Histochem 2024; 68:4004. [PMID: 38656259 PMCID: PMC11064893 DOI: 10.4081/ejh.2024.4004] [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: 03/01/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
This study investigated the impact of resveratrol on abnormal metabolic remodeling in atrial fibrillation (AF) and explored potential molecular mechanisms. An AF cell model was established by high-frequency electrical stimulation of HL-1 atrial muscle cells. Resveratrol concentrations were optimized using CCK-8 and flow cytometry. AF-induced increases in ROS and mitochondrial calcium, along with decreased adenosine triphosphate (ATP) and mitochondrial membrane potential, were observed. Resveratrol mitigated these changes and maintained normal mitochondrial morphology. Moreover, resveratrol acted through the SIRT3-dependent pathway, as evidenced by its ability to suppress AF-induced acetylation of key metabolic enzymes. SIRT3 overexpression controls acetylation modifications, suggesting its regulatory role. In conclusion, resveratrol's SIRT3-dependent pathway intervenes in AF-induced mitochondrial dysfunction, presenting a potential therapeutic avenue for AF-related metabolic disorders. This study sheds light on the role of resveratrol in mitigating AF-induced mitochondrial remodeling and highlights its potential as a novel treatment for AF.
Collapse
Affiliation(s)
- Yuejuan Cao
- Department of Cardiology, Tianjin Union Medical Center, Tianjin.
| | - Li Cui
- Department of Cardiology, Tianjin Union Medical Center, Tianjin.
| | - Shaoyong Tuo
- Department of Vascular Surgery, Tianjin Union Medical Center, Tianjin.
| | - Hongze Liu
- Department of Cardiology, Tianjin Union Medical Center, Tianjin.
| | - Shaonan Cui
- Department of Cardiology, Tianjin Union Medical Center, Tianjin.
| |
Collapse
|
10
|
Zhang Z, Zhang J, Jiao H, Tian W, Zhai X. Genetically predicted dietary macronutrient intakes and atrial fibrillation risk: a Mendelian randomization study. Eur J Med Res 2024; 29:227. [PMID: 38609963 PMCID: PMC11010414 DOI: 10.1186/s40001-024-01781-z] [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: 08/01/2023] [Accepted: 03/10/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND AND AIM Previous observational investigations have indicated a potential association between relative dietary macronutrient intakes and atrial fibrillation and flutter (AF) risk. In this study, we employed Mendelian Randomization (MR) to evaluate the presence of causality and to elucidate the specific causal relationship. METHODS We employed six, five, and three single nucleotide polymorphisms (SNPs) as instrumental variables for relative carbohydrate, protein, and fat intake, identified from a genome-wide association study that included 268,922 individuals of European descent. Furthermore, we acquired summary statistics for genome-wide association studies on AF from the FinnGen consortium, which involved 22,068 cases and 116,926 controls. To evaluate the causal estimates, we utilized the random effect inverse variance weighted method (IVW) and several other MR methods, including MR-Egger, weighted median, and MR-PRESSO, to confirm the robustness of our findings. RESULTS Our analysis indicates a convincing causal relationship between genetically predicted relative carbohydrate and protein intake and reduced AF risk. Inverse variance weighted analysis results for carbohydrates (OR = 0.29; 95% CI (0.14, 0.59); P < 0.001) and protein (OR = 0.47; 95% CI (0.26, 0.85); P = 0.01) support this association. Our MR analysis did not identify a significant causal relationship between relative fat intake and AF risk. CONCLUSION Our study provides evidence supporting a causal relationship between higher relative protein and carbohydrate intake and a lower risk of atrial fibrillation (AF).
Collapse
Affiliation(s)
- Zhuoya Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiale Zhang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Haoyang Jiao
- Institute of Documentation, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei Tian
- Gaoyang County Hospital, Baoding, 071599, Hebei Province, China.
| | - Xu Zhai
- Guanganmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
11
|
Vardas EP, Theofilis P, Oikonomou E, Vardas PE, Tousoulis D. MicroRNAs in Atrial Fibrillation: Mechanisms, Vascular Implications, and Therapeutic Potential. Biomedicines 2024; 12:811. [PMID: 38672166 PMCID: PMC11048414 DOI: 10.3390/biomedicines12040811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Atrial fibrillation (AFib), the most prevalent arrhythmia in clinical practice, presents a growing global health concern, particularly with the aging population, as it is associated with devastating complications and an impaired quality of life. Its pathophysiology is multifactorial, including the pathways of fibrosis, inflammation, and oxidative stress. MicroRNAs (miRNAs), small non-coding RNA molecules, have emerged as substantial contributors in AFib pathophysiology, by affecting those pathways. In this review, we explore the intricate relationship between miRNAs and the aforementioned aspects of AFib, shedding light on the molecular pathways as well as the potential diagnostic applications. Recent evidence also suggests a possible role of miRNA therapeutics in maintenance of sinus rhythm via the antagonism of miR-1 and miR-328, or the pharmacological upregulation of miR-27b and miR-223-3p. Unraveling the crosstalk between specific miRNA profiles and genetic predispositions may pave the way for personalized therapeutic approaches, setting the tone for precision medicine in atrial fibrillation.
Collapse
Affiliation(s)
- Emmanouil P. Vardas
- 1st Cardiology Department, General Hospital of Athens “Hippokration”, University of Athens Medical School, 11528 Athens, Greece; (E.P.V.); (P.T.)
- Department of Cardiology, General Hospital of Athens “G. Gennimatas”, 11527 Athens, Greece
| | - Panagiotis Theofilis
- 1st Cardiology Department, General Hospital of Athens “Hippokration”, University of Athens Medical School, 11528 Athens, Greece; (E.P.V.); (P.T.)
| | - Evangelos Oikonomou
- 3rd Cardiology Department, Sotiria Regional Hospital for Chest Diseases, University of Athens Medical School, 11527 Athens, Greece;
| | - Panos E. Vardas
- Biomedical Research Foundation Academy of Athens, Heart Sector, Hygeia Hospitals Group, Attica, 15123 Athens, Greece;
| | - Dimitris Tousoulis
- 1st Cardiology Department, General Hospital of Athens “Hippokration”, University of Athens Medical School, 11528 Athens, Greece; (E.P.V.); (P.T.)
| |
Collapse
|
12
|
Zhou JB, Qian LL, Wu D, Wang RX. The Role of Ferroptosis in Atrial Fibrillation: A Promising Future. Rev Cardiovasc Med 2024; 25:127. [PMID: 39076535 PMCID: PMC11264045 DOI: 10.31083/j.rcm2504127] [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: 10/08/2023] [Revised: 11/30/2023] [Accepted: 12/08/2023] [Indexed: 07/31/2024] Open
Abstract
Atrial fibrillation (AF) is one of the most common cardiac arrhythmias, with its diagnosis being closely tied to higher rates of cardiovascular morbidity and mortality. AF is associated with a range of dangerous complications including stroke and heart failure, making it a key driver of healthcare spending and a major threat to global public health. The precise mechanisms that govern AF incidence and the onset of related complications, however, remain uncertain. Ferroptotic cell death has been the focus of rising interest in the cardiac arrhythmias, and there is recent evidence supporting a role for atrial ferroptosis as a mediator of AF development. Interventional strategies focused on ferroptotic activity, such as novel ferroptosis inhibitors, have also shown promise as a means of protecting against AF through their ability to reduce iron overload. In this review, we provide a summary of the proposed mechanisms whereby ferroptosis contributes to the pathophysiology of AF and their therapeutic implications.
Collapse
Affiliation(s)
- Jia-Bin Zhou
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing
Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical
University, 214023 Wuxi, Jiangsu, China
| | - Ling-Ling Qian
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing
Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical
University, 214023 Wuxi, Jiangsu, China
| | - Dan Wu
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing
Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical
University, 214023 Wuxi, Jiangsu, China
| | - Ru-Xing Wang
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing
Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical
University, 214023 Wuxi, Jiangsu, China
| |
Collapse
|
13
|
Subati T, Yang Z, Murphy MB, Stark JM, Trykall DZ, Davies SS, Barnett JV, Murray KT. Isolevuglandins Promote Mitochondrial Dysfunction and Electrophysiologic Abnormalities in Atrial Cardiomyocytes. Cells 2024; 13:483. [PMID: 38534327 PMCID: PMC10969716 DOI: 10.3390/cells13060483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/01/2024] [Accepted: 03/02/2024] [Indexed: 03/28/2024] Open
Abstract
Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia, yet the cellular and molecular mechanisms underlying the AF substrate remain unclear. Isolevuglandins (IsoLGs) are highly reactive lipid dicarbonyl products that mediate oxidative stress-related injury. In murine hypertension, the lipid dicarbonyl scavenger 2-hydroxybenzylamine (2-HOBA) reduced IsoLGs and AF susceptibility. We hypothesized that IsoLGs mediate detrimental pathophysiologic effects in atrial cardiomyocytes that promote the AF substrate. Using Seahorse XFp extracellular flux analysis and a luminescence assay, IsoLG exposure suppressed intracellular ATP production in atrial HL-1 cardiomyocytes. IsoLGs caused mitochondrial dysfunction, with reduced mitochondrial membrane potential, increased mitochondrial reactive oxygen species (ROS) with protein carbonylation, and mitochondrial DNA damage. Moreover, they generated cytosolic preamyloid oligomers previously shown to cause similar detrimental effects in atrial cells. In mouse atrial and HL-1 cells, patch clamp experiments demonstrated that IsoLGs rapidly altered action potentials (AP), implying a direct effect independent of oligomer formation by reducing the maximum Phase 0 upstroke slope and shortening AP duration due to ionic current modifications. IsoLG-mediated mitochondrial and electrophysiologic abnormalities were blunted or totally prevented by 2-HOBA. These findings identify IsoLGs as novel mediators of oxidative stress-dependent atrial pathophysiology and support the investigation of dicarbonyl scavengers as a novel therapeutic approach to prevent AF.
Collapse
Affiliation(s)
- Tuerdi Subati
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (T.S.); (Z.Y.); (M.B.M.); (J.M.S.); (D.Z.T.); (S.S.D.); (J.V.B.)
| | - Zhenjiang Yang
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (T.S.); (Z.Y.); (M.B.M.); (J.M.S.); (D.Z.T.); (S.S.D.); (J.V.B.)
| | - Matthew B. Murphy
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (T.S.); (Z.Y.); (M.B.M.); (J.M.S.); (D.Z.T.); (S.S.D.); (J.V.B.)
| | - Joshua M. Stark
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (T.S.); (Z.Y.); (M.B.M.); (J.M.S.); (D.Z.T.); (S.S.D.); (J.V.B.)
| | - David Z. Trykall
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (T.S.); (Z.Y.); (M.B.M.); (J.M.S.); (D.Z.T.); (S.S.D.); (J.V.B.)
| | - Sean S. Davies
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (T.S.); (Z.Y.); (M.B.M.); (J.M.S.); (D.Z.T.); (S.S.D.); (J.V.B.)
| | - Joey V. Barnett
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (T.S.); (Z.Y.); (M.B.M.); (J.M.S.); (D.Z.T.); (S.S.D.); (J.V.B.)
| | - Katherine T. Murray
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (T.S.); (Z.Y.); (M.B.M.); (J.M.S.); (D.Z.T.); (S.S.D.); (J.V.B.)
- Division of Clinical Pharmacology Room 559, Preston Research Building, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| |
Collapse
|
14
|
Menezes Júnior ADS, de França-e-Silva ALG, de Oliveira JM, da Silva DM. Developing Pharmacological Therapies for Atrial Fibrillation Targeting Mitochondrial Dysfunction and Oxidative Stress: A Scoping Review. Int J Mol Sci 2023; 25:535. [PMID: 38203704 PMCID: PMC10779389 DOI: 10.3390/ijms25010535] [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: 11/18/2023] [Revised: 12/23/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Atrial fibrillation (AF) is a cardiac arrhythmia caused by electrophysiological anomalies in the atrial tissue, tissue degradation, structural abnormalities, and comorbidities. A direct relationship exists between AF and altered mitochondrial activity resulting from membrane potential loss, contractile dysfunction, or decreased ATP levels. This review aimed to elucidate the role of mitochondrial oxidative mechanisms in AF pathophysiology, the impact of mitochondrial oxidative stress on AF initiation and perpetuation, and current therapies. This review followed the Preferred Reporting Items for Systematic Reviews and the Meta-Analysis Extension for Scoping Reviews. PubMed, Excerpta Medica Database, and Scopus were explored until June 2023 using "MESH terms". Bibliographic references to relevant papers were also included. Oxidative stress is an imbalance that causes cellular damage from excessive oxidation, resulting in conditions such as AF. An imbalance in reactive oxygen species production and elimination can cause mitochondrial damage, cellular apoptosis, and cardiovascular diseases. Oxidative stress and inflammation are intrinsically linked, and inflammatory pathways are highly correlated with the occurrence of AF. AF is an intricate cardiac condition that requires innovative therapeutic approaches. The involvement of mitochondrial oxidative stress in the pathophysiology of AF introduces novel strategies for clinical treatment.
Collapse
Affiliation(s)
- Antônio da Silva Menezes Júnior
- Internal Medicine Department, Medicine School, Federal University of Goiás, Goiânia 74605-020, GO, Brazil; (A.L.G.d.F.-e.-S.); (D.M.d.S.)
- Medicine Department, Medical and Life School, Pontifical Catholic University of Goiás, Avenida Universitária, 1440, Sector Universitario, Goiânia 74605-010, GO, Brazil;
| | - Ana Luísa Guedes de França-e-Silva
- Internal Medicine Department, Medicine School, Federal University of Goiás, Goiânia 74605-020, GO, Brazil; (A.L.G.d.F.-e.-S.); (D.M.d.S.)
| | - Joyce Monteiro de Oliveira
- Medicine Department, Medical and Life School, Pontifical Catholic University of Goiás, Avenida Universitária, 1440, Sector Universitario, Goiânia 74605-010, GO, Brazil;
| | - Daniela Melo da Silva
- Internal Medicine Department, Medicine School, Federal University of Goiás, Goiânia 74605-020, GO, Brazil; (A.L.G.d.F.-e.-S.); (D.M.d.S.)
| |
Collapse
|
15
|
Pool L, Knops P, Manintveld OC, Brugts JJ, Theuns DAMJ, Brundel BJJM, de Groot NMS. The HF-AF ENERGY Trial: Nicotinamide Riboside for the Treatment of Atrial Fibrillation in Heart Failure Patients. Cardiovasc Drugs Ther 2023; 37:1243-1248. [PMID: 36227441 PMCID: PMC10721700 DOI: 10.1007/s10557-022-07382-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/10/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND The presence of atrial fibrillation (AF) in heart failure (HF) patients with reduced ejection fraction is common and associated with an increased risk of stroke, hospitalization and mortality. Recent research findings indicate that a reduction in nicotinamide adenine dinucleotide (NAD+) levels results in mitochondrial dysfunction, DNA damage and consequently cardiomyocyte impairment in experimental and clinical HF and AF. The HF-AF ENERGY trial aims to investigate the cardioprotective effects of the NAD+ precursor nicotinamide riboside (NR) treatment in ischemic heart disease patients diagnosed with AF. STUDY DESIGN The HF-AF ENERGY trial is a prospective intervention study. The study consists of a (retrospective) 4 months observation period and a 4 months intervention period. The cardioprotective effect of NR on AF burden is investigated by remote monitoring software of implantable cardiac defibrillators (ICDs), which enables continuous atrial rhythm monitoring detection. Cardiac dimension and function are examined by echocardiography. Laboratory blood analysis is performed to determine mitochondrial function markers and energy metabolism. All the study parameters are assessed at two fixed time points (pre- and post-treatment). Pre- and post-treatment outcomes are compared to determine the effects of NR treatment on AF burden, mitochondrial function markers and energy metabolism. CONCLUSION The HF-AF ENERGY trial investigates the cardioprotective effects of NR on AF burden and whether NR normalizes blood-based mitochondrial function markers and energy metabolites of the NAD metabolome in ischemic heart disease patients diagnosed with AF. The study outcomes elucidate whether NAD+ metabolism can be used as a future therapy for HF patients with AF.
Collapse
Affiliation(s)
- Lisa Pool
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Physiology, Amsterdam Cardiovascular Sciences Heart Failure and Arrhythmia, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Paul Knops
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Jasper J Brugts
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Bianca J J M Brundel
- Department of Physiology, Amsterdam Cardiovascular Sciences Heart Failure and Arrhythmia, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
16
|
Shen J, Liang J, Rejiepu M, Yuan P, Xiang J, Guo Y, Xiaokereti J, Zhang L, Tang B. Identification of a Novel Target Implicated in Chronic Obstructive Sleep Apnea-Related Atrial Fibrillation by Integrative Analysis of Transcriptome and Proteome. J Inflamm Res 2023; 16:5677-5695. [PMID: 38050561 PMCID: PMC10693830 DOI: 10.2147/jir.s438701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/21/2023] [Indexed: 12/06/2023] Open
Abstract
Objective This study aimed to identify a newly identified target involved in atrial fibrillation (AF) linked to chronic obstructive sleep apnea (COSA) through an integrative analysis of transcriptome and proteome. Methods Fifteen beagle canines were randomly assigned to three groups: control (CON), obstructive sleep apnea (OSA), and OSA with superior left ganglionated plexi ablation (OSA+GP). A COSA model was established by intermittently obstructing the endotracheal cannula during exhalation for 12 weeks. Left parasternal thoracotomy through the fourth intercostal space allowed for superior left ganglionated plexi (SLGP) ablation. In vivo open-chest electrophysiological programmed stimulation was performed to assess AF inducibility. Histological, transcriptomic, and proteomic analyses were conducted on atrial samples. Results After 12 weeks, the OSA group exhibited increased AF inducibility and longer AF durations compared to the CON group. Integrated transcriptomic and proteomic analyses identified 2422 differentially expressed genes (DEGs) and 1194 differentially expressed proteins (DEPs) between OSA and CON groups, as well as between OSA+GP and OSA groups (1850 DEGs and 1418 DEPs). The analysis revealed that differentially regulated DEGs were primarily enriched in mitochondrial biological processes in the CON-vs.-OSA and OSA-vs.-GP comparisons. Notably, the key regulatory molecule GSTZ1 was activated in OSA and inhibited by GP ablation. Conclusion These findings suggest that GSTZ1 may play a pivotal role in mitochondrial damage, triggering AF substrate formation, and increasing susceptibility to AF in the context of COSA.
Collapse
Affiliation(s)
- Jun Shen
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Junqing Liang
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Manzeremu Rejiepu
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Ping Yuan
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, People’s Republic of China
| | - Jie Xiang
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Yankai Guo
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Jiasuoer Xiaokereti
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Ling Zhang
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Baopeng Tang
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| |
Collapse
|
17
|
Benzoni P, Da Dalt L, Elia N, Popolizio V, Cospito A, Giannetti F, Dell’Era P, Olesen MS, Bucchi A, Baruscotti M, Norata GD, Barbuti A. PITX2 gain-of-function mutation associated with atrial fibrillation alters mitochondrial activity in human iPSC atrial-like cardiomyocytes. Front Physiol 2023; 14:1250951. [PMID: 38028792 PMCID: PMC10679737 DOI: 10.3389/fphys.2023.1250951] [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: 06/30/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia worldwide; however, the underlying causes of AF initiation are still poorly understood, particularly because currently available models do not allow in distinguishing the initial causes from maladaptive remodeling that induces and perpetuates AF. Lately, the genetic background has been proven to be important in the AF onset. iPSC-derived cardiomyocytes, being patient- and mutation-specific, may help solve this diatribe by showing the initial cell-autonomous changes underlying the development of the disease. Transcription factor paired-like homeodomain 2 (PITX2) has been identified as a key regulator of atrial development/differentiation, and the PITX2 genomic locus has the highest association with paroxysmal AF. PITX2 influences mitochondrial activity, and alterations in either its expression or function have been widely associated with AF. In this work, we investigate the activity of mitochondria in iPSC-derived atrial cardiomyocytes (aCMs) obtained from a young patient (24 years old) with paroxysmal AF, carrying a gain-of-function mutation in PITX2 (rs138163892) and from its isogenic control (CTRL) in which the heterozygous point mutation has been reverted to WT. PITX2 aCMs show a higher mitochondrial content, increased mitochondrial activity, and superoxide production under basal conditions when compared to CTRL aCMs. However, increasing mitochondrial workload by FCCP or β-adrenergic stimulation allows us to unmask mitochondrial defects in PITX2 aCMs, which are incapable of responding efficiently to the higher energy demand, determining ATP deficiency.
Collapse
Affiliation(s)
- Patrizia Benzoni
- The Cell Physiology MiLab, Department Biosciences, Università degli Studi di Milano, Milano, Italy
| | - Lorenzo Da Dalt
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Noemi Elia
- The Cell Physiology MiLab, Department Biosciences, Università degli Studi di Milano, Milano, Italy
- Cell Factory, Fondazione IRCCS Ca’Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Vera Popolizio
- The Cell Physiology MiLab, Department Biosciences, Università degli Studi di Milano, Milano, Italy
| | - Alessandro Cospito
- The Cell Physiology MiLab, Department Biosciences, Università degli Studi di Milano, Milano, Italy
| | - Federica Giannetti
- The Cell Physiology MiLab, Department Biosciences, Università degli Studi di Milano, Milano, Italy
- Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Istituto Auxologico Italiano IRCCS, Milano, Italy
| | - Patrizia Dell’Era
- Department of Molecular and Translational Medicine, Università degli Studi di Brescia, Brescia, Italy
| | - Morten S. Olesen
- The Heart Centre, Rigshospitalet, Laboratory for Molecular Cardiology, Department of Cardiology, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Annalisa Bucchi
- The Cell Physiology MiLab, Department Biosciences, Università degli Studi di Milano, Milano, Italy
| | - Mirko Baruscotti
- The Cell Physiology MiLab, Department Biosciences, Università degli Studi di Milano, Milano, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Andrea Barbuti
- The Cell Physiology MiLab, Department Biosciences, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
18
|
Ramos-Mondragón R, Lozhkin A, Vendrov AE, Runge MS, Isom LL, Madamanchi NR. NADPH Oxidases and Oxidative Stress in the Pathogenesis of Atrial Fibrillation. Antioxidants (Basel) 2023; 12:1833. [PMID: 37891912 PMCID: PMC10604902 DOI: 10.3390/antiox12101833] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Atrial fibrillation (AF) is the most common type of cardiac arrhythmia and its prevalence increases with age. The irregular and rapid contraction of the atria can lead to ineffective blood pumping, local blood stasis, blood clots, ischemic stroke, and heart failure. NADPH oxidases (NOX) and mitochondria are the main sources of reactive oxygen species in the heart, and dysregulated activation of NOX and mitochondrial dysfunction are associated with AF pathogenesis. NOX- and mitochondria-derived oxidative stress contribute to the onset of paroxysmal AF by inducing electrophysiological changes in atrial myocytes and structural remodeling in the atria. Because high atrial activity causes cardiac myocytes to expend extremely high energy to maintain excitation-contraction coupling during persistent AF, mitochondria, the primary energy source, undergo metabolic stress, affecting their morphology, Ca2+ handling, and ATP generation. In this review, we discuss the role of oxidative stress in activating AF-triggered activities, regulating intracellular Ca2+ handling, and functional and anatomical reentry mechanisms, all of which are associated with AF initiation, perpetuation, and progression. Changes in the extracellular matrix, inflammation, ion channel expression and function, myofibril structure, and mitochondrial function occur during the early transitional stages of AF, opening a window of opportunity to target NOX and mitochondria-derived oxidative stress using isoform-specific NOX inhibitors and mitochondrial ROS scavengers, as well as drugs that improve mitochondrial dynamics and metabolism to treat persistent AF and its transition to permanent AF.
Collapse
Affiliation(s)
- Roberto Ramos-Mondragón
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, 2301 Medical Science Research Building III, Ann Arbor, MI 48109, USA; (R.R.-M.); (L.L.I.)
| | - Andrey Lozhkin
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Aleksandr E. Vendrov
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Marschall S. Runge
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Lori L. Isom
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, 2301 Medical Science Research Building III, Ann Arbor, MI 48109, USA; (R.R.-M.); (L.L.I.)
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nageswara R. Madamanchi
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| |
Collapse
|
19
|
Kishore P, Collinet ACT, Brundel BJJM. Prevention of Atrial Fibrillation: Putting Proteostasis Derailment Back on Track. J Clin Med 2023; 12:4352. [PMID: 37445387 DOI: 10.3390/jcm12134352] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Despite the many attempts to treat atrial fibrillation (AF), the most common cardiac tachyarrhythmia in the Western world, the treatment efficacy of AF is still suboptimal. A plausible reason for the suboptimal efficacy is that the current treatments are not directed at the underlying molecular mechanisms that drive AF. Recent discoveries revealed that the derailment of specific molecular proteostasis pathways drive electrical conduction disorders, contractile dysfunction and AF. The degree of this so-called 'electropathology' corresponds to the response to anti-AF treatment. Hence, to develop effective therapies to prevent AF, understanding the molecular mechanisms is of key importance. In this review, we highlight the key modulators of proteostasis derailment and describe the mechanisms that explain how they affect electrical and contractile function in atrial cardiomyocytes and AF. The key modulators of proteostasis derailment include (1) exhaustion of cardioprotective heat shock proteins (HSPs), (2) excessive endoplasmic reticulum (ER) stress and downstream autophagic protein degradation, (3) histone deacetylase 6 (HDAC6)-induced microtubule disruption, (4) activation of DNA damage-PARP1 activation and NAD+ axis and (5) mitochondrial dysfunction. Furthermore, we discuss druggable targets within these pathways that are involved in the prevention of proteostasis derailment, as well as the targets that aid in the recovery from AF. Finally, we will elaborate on the most favorable druggable targets for (future) testing in patients with AF, as well as drugs with potential benefits for AF recovery.
Collapse
Affiliation(s)
- Preetam Kishore
- Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, The Netherlands
| | - Amelie C T Collinet
- Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, The Netherlands
| | - Bianca J J M Brundel
- Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
20
|
Bețiu AM, Noveanu L, Hâncu IM, Lascu A, Petrescu L, Maack C, Elmér E, Muntean DM. Mitochondrial Effects of Common Cardiovascular Medications: The Good, the Bad and the Mixed. Int J Mol Sci 2022; 23:13653. [PMID: 36362438 PMCID: PMC9656474 DOI: 10.3390/ijms232113653] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/20/2022] [Accepted: 10/28/2022] [Indexed: 07/25/2023] Open
Abstract
Mitochondria are central organelles in the homeostasis of the cardiovascular system via the integration of several physiological processes, such as ATP generation via oxidative phosphorylation, synthesis/exchange of metabolites, calcium sequestration, reactive oxygen species (ROS) production/buffering and control of cellular survival/death. Mitochondrial impairment has been widely recognized as a central pathomechanism of almost all cardiovascular diseases, rendering these organelles important therapeutic targets. Mitochondrial dysfunction has been reported to occur in the setting of drug-induced toxicity in several tissues and organs, including the heart. Members of the drug classes currently used in the therapeutics of cardiovascular pathologies have been reported to both support and undermine mitochondrial function. For the latter case, mitochondrial toxicity is the consequence of drug interference (direct or off-target effects) with mitochondrial respiration/energy conversion, DNA replication, ROS production and detoxification, cell death signaling and mitochondrial dynamics. The present narrative review aims to summarize the beneficial and deleterious mitochondrial effects of common cardiovascular medications as described in various experimental models and identify those for which evidence for both types of effects is available in the literature.
Collapse
Affiliation(s)
- Alina M. Bețiu
- Doctoral School Medicine-Pharmacy, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Lavinia Noveanu
- Department of Functional Sciences—Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Iasmina M. Hâncu
- Doctoral School Medicine-Pharmacy, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Ana Lascu
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- Department of Functional Sciences—Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Lucian Petrescu
- Doctoral School Medicine-Pharmacy, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, 97078 Würzburg, Germany
- Department of Internal Medicine 1, University Clinic Würzburg, 97078 Würzburg, Germany
| | - Eskil Elmér
- Mitochondrial Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, BMC A13, 221 84 Lund, Sweden
- Abliva AB, Medicon Village, 223 81 Lund, Sweden
| | - Danina M. Muntean
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- Department of Functional Sciences—Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy from Timișoara, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| |
Collapse
|
21
|
Su W, van Wijk SW, Brundel BJJM. Desmin variants: Trigger for cardiac arrhythmias? Front Cell Dev Biol 2022; 10:986718. [PMID: 36158202 PMCID: PMC9500482 DOI: 10.3389/fcell.2022.986718] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022] Open
Abstract
Desmin (DES) is a classical type III intermediate filament protein encoded by the DES gene. Desmin is abundantly expressed in cardiac, skeletal, and smooth muscle cells. In these cells, desmin interconnects several protein-protein complexes that cover cell-cell contact, intracellular organelles such as mitochondria and the nucleus, and the cytoskeletal network. The extra- and intracellular localization of the desmin network reveals its crucial role in maintaining the structural and mechanical integrity of cells. In the heart, desmin is present in specific structures of the cardiac conduction system including the sinoatrial node, atrioventricular node, and His-Purkinje system. Genetic variations and loss of desmin drive a variety of conditions, so-called desminopathies, which include desmin-related cardiomyopathy, conduction system-related atrial and ventricular arrhythmias, and sudden cardiac death. The severe cardiac disease outcomes emphasize the clinical need to understand the molecular and cellular role of desmin driving desminopathies. As the role of desmin in cardiomyopathies has been discussed thoroughly, the current review is focused on the role of desmin impairment as a trigger for cardiac arrhythmias. Here, the molecular and cellular mechanisms of desmin to underlie a healthy cardiac conduction system and how impaired desmin triggers cardiac arrhythmias, including atrial fibrillation, are discussed. Furthermore, an overview of available (genetic) desmin model systems for experimental cardiac arrhythmia studies is provided. Finally, potential implications for future clinical treatments of cardiac arrhythmias directed at desmin are highlighted.
Collapse
Affiliation(s)
- Wei Su
- Physiology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Stan W. van Wijk
- Physiology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Bianca J. J. M. Brundel
- Physiology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- *Correspondence: Bianca J. J. M. Brundel,
| |
Collapse
|
22
|
Cao S, Han C, Xuan C, Li X, Wen J, Xu D. Effects of cGMP/Akt/GSK-3β signaling pathway on atrial natriuretic peptide secretion in rabbits with rapid atrial pacing. Front Physiol 2022; 13:861981. [PMID: 36060704 PMCID: PMC9437264 DOI: 10.3389/fphys.2022.861981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Atrial natriuretic peptide (ANP) plays a pivotal role in the regulation of the cardiovascular system. The ANP level increases during atrial fibrillation (AF), suggesting that AF may provoke ANP secretion, but its potential mechanism is still unclear. In the present study, the potential mechanisms of rapid atrial pacing (RAP) regulating ANP secretion was explored. Rabbits were subjected to burst RAP, ANP secretion increased whereas cyclic guanosine monophosphate (cGMP) concentrations decreased during RAP. The p-Akt and p-GSK-3β levels decreased in atrial tissues. Natriuretic peptide receptor A (NPR-A) protein and particulate guanylate cyclase (PGC) activity were detected. The sensitivity of NPR-A to ANP decreased, leading to the decrease of PGC activity. Also, the isolated atrial perfusion system were made in the rabbit model, cGMP was shown to inhibit ANP secretion, and the Akt inhibitor LY294002 (LY) and GSK-3β inhibitor SB216763 (SB) attenuated the inhibitory effects of cGMP on ANP secretion and enhanced the inhibitory effects of cGMP on atrial dynamics. In conclusion, NPR-A interacts with ANP to regulate PGC expression, and influence the expression of cGMP during RAP, which involves in the Akt/GSK-3β signaling pathway. From the aforementioned points we conclude that cGMP regulates ANP secretion by the Akt/GSK-3β signaling pathway during atrial pacing.
Collapse
Affiliation(s)
- Shuxia Cao
- Center of Morphological Experiment, Medical College of Yanbian University, Jilin, China
| | - Chengyong Han
- Center of Morphological Experiment, Medical College of Yanbian University, Jilin, China
| | - Chunhua Xuan
- Department of Cardiology, Affiliated Hospital of Yanbian University, Jilin, China
| | - Xiangdan Li
- Center of Morphological Experiment, Medical College of Yanbian University, Jilin, China
| | - Jing Wen
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- *Correspondence: Jing Wen, ; Dongyuan Xu,
| | - Dongyuan Xu
- Center of Morphological Experiment, Medical College of Yanbian University, Jilin, China
- *Correspondence: Jing Wen, ; Dongyuan Xu,
| |
Collapse
|
23
|
Jing Y, Yang R, Chen W, Ye Q. Anti-Arrhythmic Effects of Sodium-Glucose Co-Transporter 2 Inhibitors. Front Pharmacol 2022; 13:898718. [PMID: 35814223 PMCID: PMC9263384 DOI: 10.3389/fphar.2022.898718] [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: 03/17/2022] [Accepted: 05/04/2022] [Indexed: 12/11/2022] Open
Abstract
Arrhythmias are clinically prevalent with a high mortality rate. They impose a huge economic burden, thereby substantially affecting the quality of life. Sodium-glucose co-transporter 2 inhibitor (SGLT2i) is a new type of hypoglycemic drug, which can regulate blood glucose level safely and effectively. Additionally, it reduces the occurrence and progression of heart failure and cardiovascular events significantly. Recently, studies have found that SGLT2i can alleviate the occurrence and progression of cardiac arrhythmias; however, the exact mechanism remains unclear. In this review, we aimed to discuss and summarize new literature on different modes in which SGLT2i ameliorates the occurrence and development of cardiac arrhythmias.
Collapse
|
24
|
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia despite substantial efforts to understand the pathophysiology of the condition and develop improved treatments. Identifying the underlying causative mechanisms of AF in individual patients is difficult and the efficacy of current therapies is suboptimal. Consequently, the incidence of AF is steadily rising and there is a pressing need for novel therapies. Research has revealed that defects in specific molecular pathways underlie AF pathogenesis, resulting in electrical conduction disorders that drive AF. The severity of this so-called electropathology correlates with the stage of AF disease progression and determines the response to AF treatment. Therefore, unravelling the molecular mechanisms underlying electropathology is expected to fuel the development of innovative personalized diagnostic tools and mechanism-based therapies. Moreover, the co-creation of AF studies with patients to implement novel diagnostic tools and therapies is a prerequisite for successful personalized AF management. Currently, various treatment modalities targeting AF-related electropathology, including lifestyle changes, pharmaceutical and nutraceutical therapy, substrate-based ablative therapy, and neuromodulation, are available to maintain sinus rhythm and might offer a novel holistic strategy to treat AF.
Collapse
Affiliation(s)
- Bianca J J M Brundel
- Department of Physiology, Amsterdam University Medical Centers, VU Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.
| | - Xun Ai
- Department of Physiology and Cell Biology, College of Medicine/Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | | | - Myrthe F Kuipers
- AFIPonline.org, Atrial Fibrillation Innovation Platform, Amsterdam, Netherlands
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | |
Collapse
|
25
|
Cheang I, Liao S, Zhu Q, Ni G, Wei C, Jia Z, Wu Y, Li X. Integrating Evidence of the Traditional Chinese Medicine Collateral Disease Theory in Prevention and Treatment of Cardiovascular Continuum. Front Pharmacol 2022; 13:867521. [PMID: 35370696 PMCID: PMC8964948 DOI: 10.3389/fphar.2022.867521] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular disease has become a major public health problem. The concept of “cardiovascular continuum” refers to the continuous process from the risk factors that lead to arteriosclerosis, vulnerable plaque rupture, myocardial infarction, arrhythmia, heart failure, and death. These characteristics of etiology and progressive development coincide with the idea of “preventing disease” in traditional Chinese medicine (TCM), which corresponds to the process of systemic intervention. With the update of the understanding via translational medicine, this article reviews the current evidence of the TCM collateral disease theory set prescriptions in both mechanical and clinical aspects, which could lead to the development of new therapeutic strategies for prevention and treatment.
Collapse
Affiliation(s)
- Iokfai Cheang
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Shengen Liao
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Qingqing Zhu
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Gehui Ni
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Cong Wei
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine, Shijiazhuang, China.,Hebei Yiling Hospital, Key Disciplines of State Administration of TCM for Collateral Disease, Shijiazhuang, China
| | - Zhenhua Jia
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine, Shijiazhuang, China.,Hebei Yiling Hospital, Key Disciplines of State Administration of TCM for Collateral Disease, Shijiazhuang, China
| | - Yiling Wu
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine, Shijiazhuang, China.,Hebei Yiling Hospital, Key Disciplines of State Administration of TCM for Collateral Disease, Shijiazhuang, China
| | - Xinli Li
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| |
Collapse
|
26
|
van Wijk SW, Su W, Wijdeveld LFJM, Ramos KS, Brundel BJJM. Cytoskeletal Protein Variants Driving Atrial Fibrillation: Potential Mechanisms of Action. Cells 2022; 11:416. [PMID: 35159226 PMCID: PMC8834312 DOI: 10.3390/cells11030416] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/15/2022] [Accepted: 01/20/2022] [Indexed: 11/16/2022] Open
Abstract
The most common clinical tachyarrhythmia, atrial fibrillation (AF), is present in 1-2% of the population. Although common risk factors, including hypertension, diabetes, and obesity, frequently underlie AF onset, it has been recognized that in 15% of the AF population, AF is familial. In these families, genome and exome sequencing techniques identified variants in the non-coding genome (i.e., variant regulatory elements), genes encoding ion channels, as well as genes encoding cytoskeletal (-associated) proteins. Cytoskeletal protein variants include variants in desmin, lamin A/C, titin, myosin heavy and light chain, junctophilin, nucleoporin, nesprin, and filamin C. These cytoskeletal protein variants have a strong association with the development of cardiomyopathy. Interestingly, AF onset is often represented as the initial manifestation of cardiac disease, sometimes even preceding cardiomyopathy by several years. Although emerging research findings reveal cytoskeletal protein variants to disrupt the cardiomyocyte structure and trigger DNA damage, exploration of the pathophysiological mechanisms of genetic AF is still in its infancy. In this review, we provide an overview of cytoskeletal (-associated) gene variants that relate to genetic AF and highlight potential pathophysiological pathways that drive this arrhythmia.
Collapse
Affiliation(s)
| | | | | | | | - Bianca J. J. M. Brundel
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.W.v.W.); (W.S.); (L.F.J.M.W.); (K.S.R.)
| |
Collapse
|
27
|
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia despite substantial efforts to understand the pathophysiology of the condition and develop improved treatments. Identifying the underlying causative mechanisms of AF in individual patients is difficult and the efficacy of current therapies is suboptimal. Consequently, the incidence of AF is steadily rising and there is a pressing need for novel therapies. Research has revealed that defects in specific molecular pathways underlie AF pathogenesis, resulting in electrical conduction disorders that drive AF. The severity of this so-called electropathology correlates with the stage of AF disease progression and determines the response to AF treatment. Therefore, unravelling the molecular mechanisms underlying electropathology is expected to fuel the development of innovative personalized diagnostic tools and mechanism-based therapies. Moreover, the co-creation of AF studies with patients to implement novel diagnostic tools and therapies is a prerequisite for successful personalized AF management. Currently, various treatment modalities targeting AF-related electropathology, including lifestyle changes, pharmaceutical and nutraceutical therapy, substrate-based ablative therapy, and neuromodulation, are available to maintain sinus rhythm and might offer a novel holistic strategy to treat AF.
Collapse
Affiliation(s)
- Bianca J. J. M. Brundel
- Department of Physiology, Amsterdam University Medical Centers, VU Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.,
| | - Xun Ai
- Department of Physiology and Cell Biology, College of Medicine/Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | | | - Myrthe F. Kuipers
- AFIPonline.org, Atrial Fibrillation Innovation Platform, Amsterdam, Netherlands
| | - Gregory Y. H. Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | |
Collapse
|