1
|
Xie Z, Chen T, Lu X, Zhao M, Chen Y, Wang X, Zhou H, Shen J, Guo J, Li Y. Proteomic biomarkers for noninvasive left atrial appendage thrombus prediction in patients with atrial fibrillation. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2025; 78:47-55. [PMID: 38729344 DOI: 10.1016/j.rec.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 04/24/2024] [Indexed: 05/12/2024]
Abstract
INTRODUCTION AND OBJECTIVES The CHA2DS2-VASc score, used to assess the risk of left atrial appendage thrombus (LAAT) formation in patients with atrial fibrillation (AF), has limited predictive value. Moreover, transesophageal echocardiography imaging, the gold standard diagnostic method to identify thrombi, is semi-invasive. Consequently, there is a need for alternative and noninvasive diagnostic methods for LAAT risk assessment. METHODS Deep proteomic analysis was conducted in plasma samples from 8 patients with nonvalvular AF, divided into thrombus and control groups (4 patients in each group) based on the presence or absence of LAAT. Biomarkers associated with LAAT were validated using an enzyme-linked immunosorbent assay in a cohort of 179 patients with available clinical, transthoracic, and transesophageal echocardiography data. Predictive models were developed to assess the improvement in LAAT identification. RESULTS The LAAT group had higher CHA2DS2-VASc scores, larger LA diameter, and lower LAA flow velocities. Deep proteomic analysis identified 30 differentially expressed proteins, including myosin light chain 4, prenylcysteine oxidase 1 (PCYOX1), and decorin as potential diagnostic biomarkers of LAAT. The model showed that PCYOX1 and decorin provided an area under the curve (AUC) of 0.970 for LAAT prediction compared with 0.672 in a model including the CHA2DS2-VASc score and LAA cauliflower morphology. The incremental value of proteomic biomarkers for LAAT in patients with nonvalvular AF was further confirmed with the net reclassification improvement and integrated discrimination improvement indices. CONCLUSIONS Protein levels of PCYOX1 and decorin improve the predictive performance for LAAT in patients with nonvalvular AF.
Collapse
Affiliation(s)
- ZhongHui Xie
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China; Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Chen
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Xu Lu
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China; Outpatient Department, The 44th Sanatorium of Retired Cadres in Haidian District, Beijing, China
| | - MaoXiang Zhao
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Yating Chen
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - XinYan Wang
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Hang Zhou
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Juan Shen
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Jun Guo
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China.
| | - Yang Li
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China.
| |
Collapse
|
2
|
Sweat ME, Pu WIT. Genetic and Molecular Underpinnings of Atrial Fibrillation. NPJ CARDIOVASCULAR HEALTH 2024; 1:35. [PMID: 39867228 PMCID: PMC11759492 DOI: 10.1038/s44325-024-00035-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 11/02/2024] [Indexed: 01/28/2025]
Abstract
Atrial fibrillation (AF), the most common sustained arrhythmia, increases stroke and heart failure risks. Here we review genes linked to AF and mechanisms by which they alter AF risk. We highlight gene expression differences between atrial and ventricular cardiomyocytes, regulatory mechanisms responsible for these differences, and their potential contribution to AF. Understanding AF mechanisms through the lens of atrial gene regulation is crucial to improving AF treatment.
Collapse
Affiliation(s)
- Mason E. Sweat
- Department of Cardiology, Boston Children’s
Hospital, Boston, MA 02115, USA
| | - WIlliam T. Pu
- Department of Cardiology, Boston Children’s
Hospital, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge,
MA 02138, USA
| |
Collapse
|
3
|
Relander A, Ruohonen I, Jaakkola S, Vasankari T, Nuotio I, Airaksinen KEJ, Kiviniemi T. Novel electrocardiographic classification for stroke prediction in atrial fibrillation patients undergoing cardioversion. Heart Rhythm 2024; 21:2407-2418. [PMID: 38677357 DOI: 10.1016/j.hrthm.2024.04.083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/17/2024] [Accepted: 04/20/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Abnormal conduction, structure, and function of the atrial myocardium predispose to atrial fibrillation (AF) and stroke. The usefulness of electrocardiographic indices in predicting stroke or systemic embolism (SSE) in patients undergoing cardioversion (CV) for AF remains unknown, especially in those at low estimated risk. OBJECTIVE We systematically evaluated the performance of various P-wave abnormalities (PWAs) in predicting SSE 30 days after CV (derivation cohort) and in the long term (validation cohort). METHODS Electrocardiograms (n = 1773) of AF patients undergoing an acute CV were manually reviewed. The 30-day post-CV data were used to derive a composite PWA variable. The electrocardiographic findings were validated by the long-term follow-up of patients with no anticoagulation. Electrocardiograms of 27 CAREBANK study patients with right atrial appendage biopsies were further analyzed for histopathologic validation. RESULTS During data derivation, the best performance was found with a combination of prolonged P-wave (≥180 ms), deflected P-wave morphology in lead II, biphasic P-waves in inferior leads, or increased P-terminal force (≥80 mm·ms) as markers for extensive PWA. In the validation cohort, 219 of 874 (25.1%) had extensive PWA. During a median follow-up of 4.9 years, there were 51 patients (5.8%) with SSE in total. In a competing risk model, PWA predicted SSE (adjusted hazard ratio, 2.1 per category; 95% CI, 1.4-3.1; P < .001). Areas under the curve for SSE at 3 years were 0.77, 0.79, and 0.86 for PWA, CHA2DS2-VASc, score, and their combination, respectively. On histologic evaluation, extensive PWA was associated with interstitial fibrosis (P = .033). CONCLUSION Novel electrocardiographic PWA classification provided additional prognostic insight in AF patients.
Collapse
Affiliation(s)
- Arto Relander
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Ilkka Ruohonen
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Samuli Jaakkola
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Tuija Vasankari
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Ilpo Nuotio
- Department of Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | | | - Tuomas Kiviniemi
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland.
| |
Collapse
|
4
|
Guo S, Zha L. Pathogenesis and Clinical Characteristics of Hereditary Arrhythmia Diseases. Genes (Basel) 2024; 15:1368. [PMID: 39596569 PMCID: PMC11593610 DOI: 10.3390/genes15111368] [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: 08/23/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
Hereditary arrhythmias, as a class of cardiac electrophysiologic abnormalities caused mainly by genetic mutations, have gradually become one of the most important causes of sudden cardiac death in recent years. With the continuous development of genetics and molecular biology techniques, the study of inherited arrhythmias has made remarkable progress in the past few decades. More and more disease-causing genes are being identified, and there have been advances in the application of genetic testing for disease screening in individuals with disease and their family members. Determining more refined disease prevention strategies and therapeutic regimens that are tailored to the genetic characteristics and molecular pathogenesis of different groups or individuals forms the basis of individualized treatment. Understanding advances in the study of inherited arrhythmias provides important clues to better understand their pathogenesis and clinical features. This article provides a review of the pathophysiologic alterations caused by genetic variants and their relationship to disease phenotypes, including mainly cardiac ion channelopathies and cardiac conduction disorders.
Collapse
Affiliation(s)
- Shuang Guo
- Department of Vascular Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China;
| | - Lingfeng Zha
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
5
|
Jiang X, Ly OT, Chen H, Zhang Z, Ibarra BA, Pavel MA, Brown GE, Sridhar A, Tofovic D, Swick A, Marszalek R, Vanoye CG, Navales F, George AL, Khetani SR, Rehman J, Gao Y, Darbar D, Saxena A. Transient titin-dependent ventricular defects during development lead to adult atrial arrhythmia and impaired contractility. iScience 2024; 27:110395. [PMID: 39100923 PMCID: PMC11296057 DOI: 10.1016/j.isci.2024.110395] [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: 03/14/2024] [Revised: 05/28/2024] [Accepted: 06/25/2024] [Indexed: 08/06/2024] Open
Abstract
Developmental causes of the most common arrhythmia, atrial fibrillation (AF), are poorly defined, with compensation potentially masking arrhythmic risk. Here, we delete 9 amino acids (Δ9) within a conserved domain of the giant protein titin's A-band in zebrafish and human-induced pluripotent stem cell-derived atrial cardiomyocytes (hiPSC-aCMs). We find that ttna Δ9/Δ9 zebrafish embryos' cardiac morphology is perturbed and accompanied by reduced functional output, but ventricular function recovers within days. Despite normal ventricular function, ttna Δ9/Δ9 adults exhibit AF and atrial myopathy, which are recapitulated in TTN Δ9/Δ9-hiPSC-aCMs. Additionally, action potential is shortened and slow delayed rectifier potassium current (I Ks) is increased due to aberrant atrial natriuretic peptide (ANP) levels. Strikingly, suppression of I Ks in both models prevents AF and improves atrial contractility. Thus, a small internal deletion in titin causes developmental abnormalities that increase the risk of AF via ion channel remodeling, with implications for patients who harbor disease-causing variants in sarcomeric proteins.
Collapse
Affiliation(s)
- Xinghang Jiang
- Department of Cell, Developmental, and Integrative Biology, UAB Heersink School of Medicine, Birmingham, AL 35233, USA
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Olivia T. Ly
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Hanna Chen
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Ziwei Zhang
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Beatriz A. Ibarra
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Mahmud A. Pavel
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Grace E. Brown
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Arvind Sridhar
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Physiology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - David Tofovic
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Medicine, Jesse Brown Veterans Administration, Chicago, IL 60612, USA
| | - Abigail Swick
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Richard Marszalek
- Department of Physiology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Carlos G. Vanoye
- Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
| | - Fritz Navales
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Alfred L. George
- Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
| | - Salman R. Khetani
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Jalees Rehman
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Biochemistry and Molecular Genetics, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Yu Gao
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Dawood Darbar
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Medicine, Jesse Brown Veterans Administration, Chicago, IL 60612, USA
| | - Ankur Saxena
- Department of Cell, Developmental, and Integrative Biology, UAB Heersink School of Medicine, Birmingham, AL 35233, USA
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
- O'Neal Comprehensive Cancer Center, Birmingham, AL 35233, USA
| |
Collapse
|
6
|
Wang L, Bu T, Wu X, Li L, Sun F, Cheng CY. Motor proteins, spermatogenesis and testis function. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:381-445. [PMID: 38960481 DOI: 10.1016/bs.apcsb.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
The role of motor proteins in supporting intracellular transports of vesicles and organelles in mammalian cells has been known for decades. On the other hand, the function of motor proteins that support spermatogenesis is also well established since the deletion of motor protein genes leads to subfertility and/or infertility. Furthermore, mutations and genetic variations of motor protein genes affect fertility in men, but also a wide range of developmental defects in humans including multiple organs besides the testis. In this review, we seek to provide a summary of microtubule and actin-dependent motor proteins based on earlier and recent findings in the field. Since these two cytoskeletons are polarized structures, different motor proteins are being used to transport cargoes to different ends of these cytoskeletons. However, their involvement in germ cell transport across the blood-testis barrier (BTB) and the epithelium of the seminiferous tubules remains relatively unknown. It is based on recent findings in the field, we have provided a hypothetical model by which motor proteins are being used to support germ cell transport across the BTB and the seminiferous epithelium during the epithelial cycle of spermatogenesis. In our discussion, we have highlighted the areas of research that deserve attention to bridge the gap of research in relating the function of motor proteins to spermatogenesis.
Collapse
Affiliation(s)
- Lingling Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China; Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Tiao Bu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Xiaolong Wu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Linxi Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Fei Sun
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - C Yan Cheng
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China; Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China.
| |
Collapse
|
7
|
Ryan T, Roberts JD. Stem cell models of inherited arrhythmias. NATURE CARDIOVASCULAR RESEARCH 2024; 3:420-430. [PMID: 39196215 DOI: 10.1038/s44161-024-00451-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 01/29/2024] [Indexed: 08/29/2024]
Abstract
Inherited arrhythmias are a heterogeneous group of conditions that confer risk of sudden death. Many inherited arrhythmias have been linked to pathogenic genetic variants that result in ion channel dysfunction, although current genetic testing panels fail to identify variants in many patients, potentially secondary to their underlying substrates being oligogenic or polygenic. Here we review the current state of knowledge surrounding the cellular mechanisms of inherited arrhythmias generated from stem cell models with a focus on integrating genetic and mechanistic data. The utility and limitations of human induced pluripotent stem cell models in disease modeling and drug development are also explored with a particular focus on examples of pharmacogenetics and precision medicine. We submit that progress in understanding inherited arrhythmias is likely to be made by using human induced pluripotent stem cells to model probable polygenic cases as well as to interrogate the diverse and potentially complex molecular networks implicated by genome-wide association studies.
Collapse
Affiliation(s)
- Tammy Ryan
- McMaster University, Hamilton, Ontario, Canada.
| | - Jason D Roberts
- McMaster University, Hamilton, Ontario, Canada
- Population Health Research Institute and Hamilton Health Sciences, Hamilton, Ontario, Canada
| |
Collapse
|
8
|
Vinciguerra M, Dobrev D, Nattel S. Atrial fibrillation: pathophysiology, genetic and epigenetic mechanisms. THE LANCET REGIONAL HEALTH. EUROPE 2024; 37:100785. [PMID: 38362554 PMCID: PMC10866930 DOI: 10.1016/j.lanepe.2023.100785] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/08/2023] [Accepted: 11/02/2023] [Indexed: 02/17/2024]
Abstract
Atrial fibrillation (AF) is the most common supraventricular arrhythmia affecting up to 1% of the general population. Its prevalence dramatically increases with age and could reach up to ∼10% in the elderly. The management of AF is a complex issue that is object of extensive ongoing basic and clinical research, it depends on its genetic and epigenetic causes, and it varies considerably geographically and also according to the ethnicity. Mechanistically, over the last decade, Genome Wide Association Studies have uncovered over 100 genetic loci associated with AF, and have shown that European ancestry is associated with elevated risk of AF. These AF-associated loci revolve around different types of disturbances, including inflammation, electrical abnormalities, and structural remodeling. Moreover, the discovery of epigenetic regulatory mechanisms, involving non-coding RNAs, DNA methylation and histone modification, has allowed unravelling what modifications reshape the processes leading to arrhythmias. Our review provides a current state of the field regarding the identification and functional characterization of AF-related genetic and epigenetic regulatory networks, including ethnic differences. We discuss clear and emerging connections between genetic regulation and pathophysiological mechanisms of AF.
Collapse
Affiliation(s)
- Manlio Vinciguerra
- Department of Translational Stem Cell Biology, Research Institute, Medical University of Varna, Varna, Bulgaria
- Liverpool Centre for Cardiovascular Science, Faculty of Health, Liverpool John Moores University, Liverpool, United Kingdom
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Duisburg, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Stanley Nattel
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Duisburg, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, Netherlands
- IHU LIRYC and Fondation Bordeaux Université, Bordeaux, France
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Lillo MA, Muñoz M, Rhana P, Gaul-Muller K, Quan J, Shirokova N, Xie LH, Santana LF, Fraidenraich D, Contreras JE. Remodeled connexin 43 hemichannels alter cardiac excitability and promote arrhythmias. J Gen Physiol 2023; 155:e202213150. [PMID: 37191672 PMCID: PMC10192603 DOI: 10.1085/jgp.202213150] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 01/25/2023] [Accepted: 04/24/2023] [Indexed: 05/17/2023] Open
Abstract
Connexin-43 (Cx43) is the most abundant protein forming gap junction channels (GJCs) in cardiac ventricles. In multiple cardiac pathologies, including hypertrophy and heart failure, Cx43 is found remodeled at the lateral side of the intercalated discs of ventricular cardiomyocytes. Remodeling of Cx43 has been long linked to spontaneous ventricular arrhythmia, yet the mechanisms by which arrhythmias develop are still debated. Using a model of dystrophic cardiomyopathy, we previously showed that remodeled Cx43 function as aberrant hemichannels (non-forming GJCs) that alter cardiomyocyte excitability and, consequently, promote arrhythmias. Here, we aim to evaluate if opening of remodeled Cx43 can serve as a general mechanism to alter cardiac excitability independent of cellular dysfunction associated with a particular cardiomyopathy. To address this issue, we used a genetically modified Cx43 knock-in mouse (S3A) that promotes cardiac remodeling of Cx43 protein without apparent cardiac dysfunction. Importantly, when S3A mice were subjected to cardiac stress using the β-adrenergic agonist isoproterenol (Iso), they displayed acute and severe arrhythmias, which were not observed in WT mice. Pretreatment of S3A mice with the Cx43 hemichannel blocker, Gap19, prevented Iso-induced abnormal electrocardiographic behavior. At the cellular level, when compared with WT, Iso-treated S3A cardiomyocytes showed increased membrane permeability, greater plasma membrane depolarization, and Ca2+ overload, which likely caused prolonged action potentials, delayed after depolarizations, and triggered activity. All these cellular dysfunctions were also prevented by Cx43 hemichannel blockers. Our results support the notion that opening of remodeled Cx43 hemichannels, regardless of the type of cardiomyopathy, is sufficient to mediate cardiac-stress-induced arrhythmogenicity.
Collapse
Affiliation(s)
- Mauricio A. Lillo
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University, New Jersey Medical School, Newark, NJ, USA
| | - Manuel Muñoz
- Department of Physiology and Membrane Biology, University of California, Davis. Davis, CA, USA
| | - Paula Rhana
- Department of Physiology and Membrane Biology, University of California, Davis. Davis, CA, USA
| | - Kelli Gaul-Muller
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University, New Jersey Medical School, Newark, NJ, USA
| | - Jonathan Quan
- Department of Physiology and Membrane Biology, University of California, Davis. Davis, CA, USA
| | - Natalia Shirokova
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University, New Jersey Medical School, Newark, NJ, USA
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, Rutgers University, New Jersey Medical School, Newark, NJ, USA
| | - Luis Fernando Santana
- Department of Physiology and Membrane Biology, University of California, Davis. Davis, CA, USA
| | - Diego Fraidenraich
- Department of Cell Biology and Molecular Medicine, Rutgers University, New Jersey Medical School, Newark, NJ, USA
| | - Jorge E. Contreras
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University, New Jersey Medical School, Newark, NJ, USA
- Department of Physiology and Membrane Biology, University of California, Davis. Davis, CA, USA
| |
Collapse
|
10
|
Xu X, Yu Z, Ai N, Liufu S, Liu X, Chen B, Li X, Jiang J, Zhang Y, Ma H, Yin Y. Molecular Mechanism of MYL4 Regulation of Skeletal Muscle Development in Pigs. Genes (Basel) 2023; 14:1267. [PMID: 37372447 DOI: 10.3390/genes14061267] [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: 05/17/2023] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The processes of muscle growth and development, including myoblast proliferation, migration, differentiation, and fusion, are modified by a variety of regulatory factors. MYL4 plays an important role in atrial development, atrial cardiomyopathy, muscle-fiber size, and muscle development. The structural variation (SV) of MYL4 was found via the de novo sequencing of Ningxiang pigs, and the existence of SV was verified in the experiments. The genotype distribution of Ningxiang pigs and Large White pigs was detected, and it was found that Ningxiang pigs were mainly of the BB genotype and that Large White pigs were mainly of the AB genotype. However, the molecular mechanisms behind the MYL4-mediated regulation of skeletal muscle development need to be deeply explored. Therefore, RT-qPCR, 3'RACE, CCK8, EdU, Western blot, immunofluorescence, flow cytometry, and bioinformation analysis were used to explore the function of MYL4 in myoblast development. The cDNA of MYL4 was successfully cloned from Ningxiang pigs, and its physicochemical properties were predicted. The expression profiles in six tissues and four stages of Ningxiang pigs and Large White pigs were found to be the highest in the lungs and 30 days after birth. The expression of MYL4 increased gradually with the extension of the myogenic differentiation time. The myoblast function test showed that the overexpression of MYL4 inhibited proliferation and promoted apoptosis and differentiation. The knockdown of MYL4 showed the opposite result. These results enhance our understanding of the molecular mechanisms of muscle development and provide a solid theoretical foundation for further exploring the role of the MYL4 gene in muscle development.
Collapse
Affiliation(s)
- Xueli Xu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Zonggang Yu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Nini Ai
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Sui Liufu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Xiaolin Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Bohe Chen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Xintong Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Jun Jiang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Yuebo Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Haiming Ma
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Yulong Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| |
Collapse
|
11
|
Zhong Y, Tang K, Nattel S, Zhai M, Gong S, Yu Q, Zeng Y, E G, Maimaitiaili N, Wang J, Xu Y, Peng W, Li H. Myosin light-chain 4 gene-transfer attenuates atrial fibrosis while correcting autophagic flux dysregulation. Redox Biol 2023; 60:102606. [PMID: 36645977 PMCID: PMC9860351 DOI: 10.1016/j.redox.2023.102606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVES To determine the role of MYL4 regulation of lysosomal function and its disturbance in fibrotic atrial cardiomyopathy. BACKGROUND We have previously demonstrated that the atrial-specific essential light chain protein MYL4 is required for atrial contractile, electrical, and structural integrity. MYL4 mutation/dysfunction leads to atrial fibrosis, standstill, and dysrhythmia. However, the underlying pathogenic mechanisms remain unclear. METHODS AND RESULTS Rats subjected to knock-in of a pathogenic MYL4 mutant (p.E11K) developed fibrotic atrial cardiomyopathy. Proteome analysis and single-cell RNA sequencing indicate enrichment of autophagy pathways in mutant-MYL4 atrial dysfunction. Immunofluorescence and electron microscopy revealed undegraded autophagic vesicles accumulated in MYL4p.E11K rat atrium. Next, we identified that dysfunctional MYL4 protein impairs autophagy flux in vitro and in vivo. Cardiac lysosome positioning and mobility were regulated by MYL4 in cardiomyocytes, which affected lysosomal acidification and maturation of lysosomal cathepsins. We then examined the effects of MYL4 overexpression via adenoviral gene-transfer on atrial cardiomyopathy induced by MYL4 mutation: MYL4 protein overexpression attenuated atrial structural remodeling and autophagy dysfunction. CONCLUSIONS MYL4 regulates autophagic flux in atrial cardiomyocytes via lysosomal mobility. MYL4 overexpression attenuates MYL4 p.E11K induced fibrotic atrial cardiomyopathy, while correcting autophagy and lysosomal function. These results provide a molecular basis for MYL4-mutant induced fibrotic atrial cardiomyopathy and identify a potential biological-therapy approach for the treatment of atrial fibrosis.
Collapse
Affiliation(s)
- Yuan Zhong
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kai Tang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Stanley Nattel
- Department of Medicine, Montreal Heart Institute, Montreal, Quebec, Canada; Université de Montréal, Quebec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany; HIU LYRIC and Fondation Bordeaux Université de Bordeaux, France
| | - Ming Zhai
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shiyu Gong
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qing Yu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanxi Zeng
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guangxi E
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Nuerbiyemu Maimaitiaili
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Wenhui Peng
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Hailing Li
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
12
|
Leybaert L, De Smet MA, Lissoni A, Allewaert R, Roderick HL, Bultynck G, Delmar M, Sipido KR, Witschas K. Connexin hemichannels as candidate targets for cardioprotective and anti-arrhythmic treatments. J Clin Invest 2023; 133:168117. [PMID: 36919695 PMCID: PMC10014111 DOI: 10.1172/jci168117] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023] Open
Abstract
Connexins are crucial cardiac proteins that form hemichannels and gap junctions. Gap junctions are responsible for the propagation of electrical and chemical signals between myocardial cells and cells of the specialized conduction system in order to synchronize the cardiac cycle and steer cardiac pump function. Gap junctions are normally open, while hemichannels are closed, but pathological circumstances may close gap junctions and open hemichannels, thereby perturbing cardiac function and homeostasis. Current evidence demonstrates an emerging role of hemichannels in myocardial ischemia and arrhythmia, and tools are now available to selectively inhibit hemichannels without inhibiting gap junctions as well as to stimulate hemichannel incorporation into gap junctions. We review available experimental evidence for hemichannel contributions to cellular pro-arrhythmic events in ventricular and atrial cardiomyocytes, and link these to insights at the level of molecular control of connexin-43-based hemichannel opening. We conclude that a double-edged approach of both preventing hemichannel opening and preserving gap junctional function will be key for further research and development of new connexin-based experimental approaches for treating heart disease.
Collapse
Affiliation(s)
- Luc Leybaert
- Physiology Group, Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Maarten Aj De Smet
- Physiology Group, Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Alessio Lissoni
- Physiology Group, Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Rosalie Allewaert
- Physiology Group, Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - H Llewelyn Roderick
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, and
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Mario Delmar
- Leon H. Charney Division of Cardiology, School of Medicine, New York University, New York, USA
| | - Karin R Sipido
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, and
| | - Katja Witschas
- Physiology Group, Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW Atrial fibrillation is the most common cardiac arrhythmia worldwide. There is considerable interest in better understanding the molecular genetics and biology of atrial fibrillation to inform the development of new therapies and improve clinical management. This review summarizes recent advances in our understanding of the genetic basis of atrial fibrillation and new efforts to utilize genetics to inform clinical management. RECENT FINDINGS Genome-wide association studies in diverse populations have increased the number of genetic loci associated with atrial fibrillation and its specific subtypes. Large-scale biobanks with deep phenotyping have provided invaluable data to study the impact of both common and rare variants on atrial fibrillation, susceptibility, and prognosis. Polygenic risk scores help improve individual atrial fibrillation risk stratification and prognostication. SUMMARY Our understanding of atrial fibrillation genetics is rapidly improving with larger and more diverse genome-wide association studies. Translating genetic discoveries into molecular pathways and new therapeutic targets remains a bottleneck in the development of new therapies for atrial fibrillation. Genetic risk scores have shown early promise in improving atrial fibrillation risk stratification; however, their broader utility for the general population remains unclear.
Collapse
Affiliation(s)
- David S M Lee
- Medical Scientist Training Program, University of Pennsylvania Perelman School of Medicine
| | - Scott M Damrauer
- Corporal Michael J. Crescenz VA Medical Center.,Department of Surgery.,Department of Genetics, University of Pennsylvania Perelman School of Medicine
| | - Michael G Levin
- Corporal Michael J. Crescenz VA Medical Center.,Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
MacRae CA, Peterson RT. Zebrafish as a Mainstream Model for In Vivo Systems Pharmacology and Toxicology. Annu Rev Pharmacol Toxicol 2023; 63:43-64. [PMID: 36151053 DOI: 10.1146/annurev-pharmtox-051421-105617] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Pharmacology and toxicology are part of a much broader effort to understand the relationship between chemistry and biology. While biomedicine has necessarily focused on specific cases, typically of direct human relevance, there are real advantages in pursuing more systematic approaches to characterizing how health and disease are influenced by small molecules and other interventions. In this context, the zebrafish is now established as the representative screenable vertebrate and, through ongoing advances in the available scale of genome editing and automated phenotyping, is beginning to address systems-level solutions to some biomedical problems. The addition of broader efforts to integrate information content across preclinical model organisms and the incorporation of rigorous analytics, including closed-loop deep learning, will facilitate efforts to create systems pharmacology and toxicology with the ability to continuously optimize chemical biological interactions around societal needs. In this review, we outline progress toward this goal.
Collapse
Affiliation(s)
- Calum A MacRae
- Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA;
| | | |
Collapse
|
15
|
Hu H, Wang L, Li H, Li H, Chen X, Peng W, Wang J, Zhao Y, Liu M, Li D. Long-term amelioration of an early-onset familial atrial fibrillation model with AAV-mediated in vivo gene therapy. FUNDAMENTAL RESEARCH 2022; 2:829-835. [PMID: 38933375 PMCID: PMC11197581 DOI: 10.1016/j.fmre.2022.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/05/2022] [Accepted: 05/05/2022] [Indexed: 10/18/2022] Open
Abstract
Atrial fibrillation (AF) is a common cardiac disease with high prevalence in the general population. Despite a mild manifestation at the onset stage, it causes serious consequences, including sudden death, when the disease progresses to the late stage. Most available treatments of AF focus on symptom management or alleviation, due to a lack of fundamental knowledge and the fact that considerable variations of AF exist. With the popularisation of the next-generation sequencing technology, several causal genetic factors, including MYL4, have been discovered to contribute to AF, giving hope to developing its gene therapies. In this study, we attempted to treat a previously established rat AF model, which carried Myl4E11K/E11K loss of function mutation, via overexpression of exogenous wild-type Myl4 by AAV9 vectors. Our results showed that delivery of Myl4 expressing AAV9 to postnatal rat models rescued the symptoms of AF, indicating the therapeutic potential that early gene therapy intervention can achieve long-term effects in treating cardiac arrhythmias caused by gene mutations.
Collapse
Affiliation(s)
- Handan Hu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Liren Wang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Huiying Li
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
- Southern Medical University Affiliated Fengxian Hospital, Shanghai 201499, China
| | - Hailing Li
- The Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Xi Chen
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Wenhui Peng
- The Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Yongxiang Zhao
- National Center for International Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Theranostics, Guangxi Medical University, Guangxi 530021, China
| | - Mingyao Liu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Dali Li
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
16
|
Relationship between Serum miR-106 and MYL4 Levels and the Prevalence, Risk Stratification, and Prognosis of Atrial Fibrillation. J Immunol Res 2022; 2022:1069866. [PMID: 35874900 PMCID: PMC9303158 DOI: 10.1155/2022/1069866] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/16/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
Objective To analyze the predictive value of serum microRNA-106 (miRNA-106), miR-106, and myosin light chain 4 (MYL4) levels on the prevalence of atrial fibrillation and to explore the relationship between serum miR-106 and MYL4 and the risk stratification and prognosis of atrial fibrillation, thereby providing basis for them to become clinical targets for the treatment of atrial fibrillation in the future. Methods 300 patients with atrial fibrillation treated in our hospital from May 2017 to March 2019 were selected as the atrial fibrillation group, and 300 healthy people who came to our hospital for physical examination in the same period were selected as the control group. The general data of the subjects in the two groups were collected. The serum miR-106 level of the subjects in the two groups was detected by fluorescence quantitative polymerase chain reaction (PCR), and the level of MYL4 was detected by enzyme-linked immunosorbent assay (ELISA). The expression of miR-106 and MYL4 in the myocardium was observed by immunohistochemistry. The relationship between the levels of serum miR-106 and MYL4 and the prevalence of atrial fibrillation and the score of atrial fibrillation thromboembolism risk stratification scoring system (cha2ds2) was compared between the two groups. The relationship between serum level of miR-106 and prognosis of patients with atrial fibrillation was analyzed. Results The systolic blood pressure, diastolic blood pressure, total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), and left anterior descending artery (LAD) in the atrial fibrillation group were significantly higher than those in the control group, while HDL-C and left ventricular ejection fraction (LVEF) were significantly lower than those in the control group (P < 0.01). The level of serum miR-106 in patients with atrial fibrillation was significantly higher than that in the control group, whereas the level of MYL4 was significantly lower than that in the control group (P < 0.01). miR-106 was mainly localized in the cytoplasm, and the positive expression rate of miR-106 was 71.43% (81/115) in patients with atrial fibrillation and 21.74% (25/115) in patients with sinus rhythm. MYL4 was mainly located in the cell membrane and the positive expression rate of MYL4 was 24.35% (28/115) in patients with atrial fibrillation and 64.35% (74/115) in patients with sinus rhythm. With the increase of the severity of atrial fibrillation, the level of serum miR-106 gradually increased and the level of MYL4 gradually decreased, which were statistically significant compared with the control group (P < 0.05). With the increase of miR-106 level and the decrease of MYL4 level, the prevalence of atrial fibrillation gradually increased. With the increase of cha2ds2 score, the level of serum miR-106 increased and the level of MYL4 decreased. The survival rate of patients with miR − 106 ≤ 1.96 was significantly higher than that of patients with miR − 106 > 1.96. The survival rate of patients with MYL4 ≥ 0.24 was significantly higher than that of patients with MYL4 < 0.24. At the same time, TC and LDL-C were included in the analysis. The results showed that the survival rate of patients with TC ≤ 4.5 mmol/L was significantly higher than that of patients with TC > 4.5 mmol/L, and that of patients with LDL-C ≤ 2.6 mmol/L was significantly higher than that of patients with LDL-C > 2.6 mmol/L. Conclusion Serum miR-106 and MYL4 levels are closely related to the prevalence of atrial fibrillation, which can reflect the risk of thromboembolism in patients with atrial fibrillation and can be used as a biological indicator to predict the prognosis of patients with atrial fibrillation.
Collapse
|
17
|
Liu Y, Dai M, Yang P, Cao L, Lu L. Src-homology domain 2 containing protein tyrosine phosphatase-1 (SHP-1) directly binds to proto-oncogene tyrosine-protein kinase Src (c-Src) and promotes the transcriptional activation of connexin 43 (Cx43). Bioengineered 2022; 13:13534-13543. [PMID: 35659197 PMCID: PMC9276044 DOI: 10.1080/21655979.2022.2079252] [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] [Indexed: 11/09/2022] Open
Abstract
The prevalence of atrial fibrillation (AF), which is one of the common arrhythmias in clinics, is increasing sharply and has affected millions of patients, which is expected to triple by 2050. The purpose of the study was to explore the regulatory relationship between Src-homology domain 2 containing protein tyrosine phosphatase-1 (SHP-1) and proto-oncogene tyrosine-protein kinase Src (c-Src) and the regulation of Connexins 43 (Cx43), and its effect on AF was also studied. Mouse atrial myocyte line (HL-1 cell line) was used as the research object. After overexpression of SHP-1, the expressions of p-c-Src, Cx43, and SHP-1 were detected by Western blot and cellular immunofluorescence, respectively. The location and interaction of SHP-1 and c-Src in the cells were detected by immunofluorescence co-localization and co-immunoprecipitation (Co-IP). The regulation of c-Src and Cx43 was detected by DNA pull down, chromatin co-immunoprecipitation (CHIP), and dual-luciferase reporter system. The results revealed that overexpression of SHP-1 could inhibit the phosphorylation and activation of c-Src and increase the expression of Cx43. Moreover, there was a direct binding between SHP-1 and c-Src, and c-Src could bind to the promoter region of Cx43 and inhibit the transcription of Cx43. In conclusion, SHP-1 could bind to c-Src and inhibit the activity of c-Src, thus enhancing the transcriptional activation of Cx43 and improving the function of gap junction.
Collapse
Affiliation(s)
- YiHao Liu
- Department of Cardiovascular Medicine, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Meng Dai
- Department of Palliative Medicine, Chongqing University Cancer Hospital, Chongqing, China
| | - PengHui Yang
- Department of Cardiovascular Medicine, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Li Cao
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Lu
- Department of Critical Care Medicine, University-Town Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
18
|
Ly OT, Chen H, Brown GE, Hong L, Wang X, Han YD, Pavel MA, Sridhar A, Maienschein-Cline M, Chalazan B, Ong SG, Abdelhady K, Massad M, Rizkallah LE, Rehman J, Khetani SR, Darbar D. Mutant ANP induces mitochondrial and ion channel remodeling in a human iPSC-derived atrial fibrillation model. JCI Insight 2022; 7:155640. [PMID: 35393944 PMCID: PMC9057627 DOI: 10.1172/jci.insight.155640] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/10/2022] [Indexed: 11/17/2022] Open
Abstract
Human induced pluripotent stem cell–derived cardiomyocytes (iPSC-CMs) can model heritable arrhythmias to personalize therapies for individual patients. Although atrial fibrillation (AF) is a leading cause of cardiovascular morbidity and mortality, current platforms to generate iPSC-atrial (a) CMs are inadequate for modeling AF. We applied a combinatorial engineering approach, which integrated multiple physiological cues, including metabolic conditioning and electrical stimulation, to generate mature iPSC-aCMs. Using the patient’s own atrial tissue as a gold standard benchmark, we assessed the electrophysiological, structural, metabolic, and molecular maturation of iPSC-aCMs. Unbiased transcriptomic analysis and inference from gene regulatory networks identified key gene expression pathways and transcription factors mediating atrial development and maturation. Only mature iPSC-aCMs generated from patients with heritable AF carrying the non-ion channel gene (NPPA) mutation showed enhanced expression and function of a cardiac potassium channel and revealed mitochondrial electron transport chain dysfunction. Collectively, we propose that ion channel remodeling in conjunction with metabolic defects created an electrophysiological substrate for AF. Overall, our electro-metabolic approach generated mature human iPSC-aCMs that unmasked the underlying mechanism of the first non-ion channel gene, NPPA, that causes AF. Our maturation approach will allow for the investigation of the molecular underpinnings of heritable AF and the development of personalized therapies.
Collapse
Affiliation(s)
- Olivia T Ly
- Division of Cardiology, Department of Medicine.,Department of Biomedical Engineering
| | - Hanna Chen
- Division of Cardiology, Department of Medicine
| | | | - Liang Hong
- Division of Cardiology, Department of Medicine
| | - Xinge Wang
- Division of Cardiology, Department of Medicine.,Department of Biomedical Engineering
| | | | | | - Arvind Sridhar
- Division of Cardiology, Department of Medicine.,Department of Physiology
| | | | | | - Sang-Ging Ong
- Division of Cardiology, Department of Medicine.,Department of Pharmacology and Regenerative Medicine; and
| | - Khaled Abdelhady
- Division of Cardiothoracic Surgery, Department of Surgery, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Malek Massad
- Division of Cardiothoracic Surgery, Department of Surgery, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Lona Ernst Rizkallah
- Division of Cardiothoracic Surgery, Department of Surgery, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Jalees Rehman
- Division of Cardiology, Department of Medicine.,Department of Biomedical Engineering.,Division of Cardiothoracic Surgery, Department of Surgery, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | - Dawood Darbar
- Division of Cardiology, Department of Medicine.,Department of Biomedical Engineering.,Department of Physiology.,Division of Cardiothoracic Surgery, Department of Surgery, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
19
|
Guo YH, Yang YQ. Atrial Fibrillation: Focus on Myocardial Connexins and Gap Junctions. BIOLOGY 2022; 11:489. [PMID: 35453689 PMCID: PMC9029470 DOI: 10.3390/biology11040489] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 06/14/2023]
Abstract
Atrial fibrillation (AF) represents the most common type of clinical cardiac arrhythmia worldwide and contributes to substantial morbidity, mortality and socioeconomic burden. Aggregating evidence highlights the strong genetic basis of AF. In addition to chromosomal abnormalities, pathogenic mutations in over 50 genes have been causally linked to AF, of which the majority encode ion channels, cardiac structural proteins, transcription factors and gap junction channels. In the heart, gap junctions comprised of connexins (Cxs) form intercellular pathways responsible for electrical coupling and rapid coordinated action potential propagation between adjacent cardiomyocytes. Among the 21 isoforms of connexins already identified in the mammal genomes, 5 isoforms (Cx37, Cx40, Cx43, Cx45 and Cx46) are expressed in human heart. Abnormal electrical coupling between cardiomyocytes caused by structural remodeling of gap junction channels (alterations in connexin distribution and protein levels) has been associated with enhanced susceptibility to AF and recent studies have revealed multiple causative mutations or polymorphisms in 4 isoforms of connexins predisposing to AF. In this review, an overview of the genetics of AF is made, with a focus on the roles of mutant myocardial connexins and gap junctions in the pathogenesis of AF, to underscore the hypothesis that cardiac connexins are a major molecular target in the management of AF.
Collapse
Affiliation(s)
- Yu-Han Guo
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China;
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China;
- Cardiovascular Research Laboratory, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
- Center Laboratory, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
| |
Collapse
|
20
|
Gauvrit S, Bossaer J, Lee J, Collins MM. Modeling Human Cardiac Arrhythmias: Insights from Zebrafish. J Cardiovasc Dev Dis 2022; 9:jcdd9010013. [PMID: 35050223 PMCID: PMC8779270 DOI: 10.3390/jcdd9010013] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/23/2021] [Accepted: 12/25/2021] [Indexed: 12/13/2022] Open
Abstract
Cardiac arrhythmia, or irregular heart rhythm, is associated with morbidity and mortality and is described as one of the most important future public health challenges. Therefore, developing new models of cardiac arrhythmia is critical for understanding disease mechanisms, determining genetic underpinnings, and developing new therapeutic strategies. In the last few decades, the zebrafish has emerged as an attractive model to reproduce in vivo human cardiac pathologies, including arrhythmias. Here, we highlight the contribution of zebrafish to the field and discuss the available cardiac arrhythmia models. Further, we outline techniques to assess potential heart rhythm defects in larval and adult zebrafish. As genetic tools in zebrafish continue to bloom, this model will be crucial for functional genomics studies and to develop personalized anti-arrhythmic therapies.
Collapse
|
21
|
Ke ZP, Zhang GF, Guo YH, Sun YM, Wang J, Li N, Qiu XB, Xu YJ, Yang YQ. A novel PRRX1 loss-of-function variation contributing to familial atrial fibrillation and congenital patent ductus arteriosus. Genet Mol Biol 2022; 45:e20210378. [PMID: 35377386 PMCID: PMC8978609 DOI: 10.1590/1678-4685-gmb-2021-0378] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/25/2022] [Indexed: 12/27/2022] Open
Abstract
Atrial fibrillation (AF) represents the most common type of sustained cardiac arrhythmia in humans and confers a significantly increased risk for thromboembolic stroke, congestive heart failure and premature death. Aggregating evidence emphasizes the predominant genetic defects underpinning AF and an increasing number of deleterious variations in more than 50 genes have been involved in the pathogenesis of AF. Nevertheless, the genetic basis underlying AF remains incompletely understood. In the current research, by whole-exome sequencing and Sanger sequencing analysis in a family with autosomal-dominant AF and congenital patent ductus arteriosus (PDA), a novel heterozygous variation in the PRRX1 gene encoding a homeobox transcription factor critical for cardiovascular development, NM_022716.4:c.373G>T;p.(Glu125*), was identified to be in co-segregation with AF and PDA in the whole family. The truncating variation was not detected in 306 unrelated healthy individuals employed as controls. Quantitative biological measurements with a reporter gene analysis system revealed that the Glu125*-mutant PRRX1 protein failed to transactivate its downstream target genes SHOX2 and ISL1, two genes that have been causally linked to AF. Conclusively, the present study firstly links PRRX1 loss-of-function variation to AF and PDA, suggesting that AF and PDA share a common abnormal developmental basis in a proportion of cases.
Collapse
Affiliation(s)
| | | | - Yu-Han Guo
- Fudan University, China; Fudan University, China
| | | | | | - Ning Li
- Shanghai Jiao Tong University, China
| | | | - Ying-Jia Xu
- Fudan University, China; Fudan University, China
| | - Yi-Qing Yang
- Fudan University, China; Fudan University, China; Fudan University, China; Fudan University, China
| |
Collapse
|
22
|
Guo XJ, Qiu XB, Wang J, Guo YH, Yang CX, Li L, Gao RF, Ke ZP, Di RM, Sun YM, Xu YJ, Yang YQ. PRRX1 Loss-of-Function Mutations Underlying Familial Atrial Fibrillation. J Am Heart Assoc 2021; 10:e023517. [PMID: 34845933 PMCID: PMC9075371 DOI: 10.1161/jaha.121.023517] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Atrial fibrillation (AF) is the most common form of clinical cardiac dysrhythmia responsible for thromboembolic cerebral stroke, congestive heart failure, and death. Aggregating evidence highlights the strong genetic basis of AF. Nevertheless, AF is of pronounced genetic heterogeneity, and in an overwhelming majority of patients, the genetic determinants underpinning AF remain elusive. Methods and Results By genome‐wide screening with polymorphic microsatellite markers and linkage analysis in a 4‐generation Chinese family affected with autosomal‐dominant AF, a novel locus for AF was mapped to chromosome 1q24.2–q25.1, a 3.20‐cM (≈4.19 Mbp) interval between markers D1S2851 and D1S218, with the greatest 2‐point logarithm of odds score of 4.8165 for the marker D1S452 at recombination fraction=0.00. Whole‐exome sequencing and bioinformatics analyses showed that within the mapping region, only the mutation in the paired related homeobox 1 (PRRX1) gene, NM_022716.4:c.319C>T;(p.Gln107*), cosegregated with AF in the family. In addition, sequencing analyses of PRRX1 in another cohort of 225 unrelated patients with AF revealed a new mutation, NM_022716.4:c.437G>T; (p.Arg146Ile), in a patient. The 2 mutations were absent in 908 control subjects. Biological analyses in HeLa cells demonstrated that the 2 mutants had significantly diminished transactivation on the target genes ISL1 and SHOX2 and markedly decreased ability to bind the promoters of ISL1 and SHOX2 (2 genes causally linked to AF), although with normal intracellular distribution. Conclusions This study first indicates that PRRX1 loss‐of‐function mutations predispose to AF, which provides novel insight into the molecular pathogenesis underpinning AF, implying potential implications for precisive prophylaxis and management of AF.
Collapse
Affiliation(s)
- Xiao-Juan Guo
- Department of Cardiology and the Center for Complex Cardiac Arrhythmias of Minhang District Shanghai Fifth People's HospitalFudan University Shanghai China
| | - Xing-Biao Qiu
- Department of Cardiology Shanghai Chest HospitalShanghai Jiao Tong University Shanghai China
| | - Jun Wang
- Department of Cardiology Shanghai Jing'an District Central HospitalFudan University Shanghai China
| | - Yu-Han Guo
- Department of Cardiology and the Center for Complex Cardiac Arrhythmias of Minhang District Shanghai Fifth People's HospitalFudan University Shanghai China
| | - Chen-Xi Yang
- Department of Cardiology and the Center for Complex Cardiac Arrhythmias of Minhang District Shanghai Fifth People's HospitalFudan University Shanghai China
| | - Li Li
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East HospitalTongji University School of Medicine Shanghai China.,Institute of Medical GeneticsTongji University Shanghai China
| | - Ri-Feng Gao
- Department of Cardiology and the Center for Complex Cardiac Arrhythmias of Minhang District Shanghai Fifth People's HospitalFudan University Shanghai China
| | - Zun-Ping Ke
- Department of Cardiology and the Center for Complex Cardiac Arrhythmias of Minhang District Shanghai Fifth People's HospitalFudan University Shanghai China
| | - Ruo-Min Di
- Department of Cardiology and the Center for Complex Cardiac Arrhythmias of Minhang District Shanghai Fifth People's HospitalFudan University Shanghai China
| | - Yu-Min Sun
- Department of Cardiology Shanghai Jing'an District Central HospitalFudan University Shanghai China
| | - Ying-Jia Xu
- Department of Cardiology and the Center for Complex Cardiac Arrhythmias of Minhang District Shanghai Fifth People's HospitalFudan University Shanghai China
| | - Yi-Qing Yang
- Department of Cardiology and the Center for Complex Cardiac Arrhythmias of Minhang District Shanghai Fifth People's HospitalFudan University Shanghai China.,Cardiovascular Research Laboratory and Central Laboratory Shanghai Fifth People's HospitalFudan University Shanghai China
| |
Collapse
|
23
|
Papathanasiou KA, Giotaki SG, Vrachatis DA, Siasos G, Lambadiari V, Iliodromitis KE, Kossyvakis C, Kaoukis A, Raisakis K, Deftereos G, Papaioannou TG, Giannopoulos G, Avramides D, Deftereos SG. Molecular Insights in Atrial Fibrillation Pathogenesis and Therapeutics: A Narrative Review. Diagnostics (Basel) 2021; 11:diagnostics11091584. [PMID: 34573926 PMCID: PMC8470040 DOI: 10.3390/diagnostics11091584] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022] Open
Abstract
The prevalence of atrial fibrillation (AF) is bound to increase globally in the following years, affecting the quality of life of millions of people, increasing mortality and morbidity, and beleaguering health care systems. Increasingly effective therapeutic options against AF are the constantly evolving electroanatomic substrate mapping systems of the left atrium (LA) and ablation catheter technologies. Yet, a prerequisite for better long-term success rates is the understanding of AF pathogenesis and maintenance. LA electrical and anatomical remodeling remains in the epicenter of current research for novel diagnostic and treatment modalities. On a molecular level, electrical remodeling lies on impaired calcium handling, enhanced inwardly rectifying potassium currents, and gap junction perturbations. In addition, a wide array of profibrotic stimuli activates fibroblast to an increased extracellular matrix turnover via various intermediaries. Concomitant dysregulation of the autonomic nervous system and the humoral function of increased epicardial adipose tissue (EAT) are established mediators in the pathophysiology of AF. Local atrial lymphomononuclear cells infiltrate and increased inflammasome activity accelerate and perpetuate arrhythmia substrate. Finally, impaired intracellular protein metabolism, excessive oxidative stress, and mitochondrial dysfunction deplete atrial cardiomyocyte ATP and promote arrhythmogenesis. These overlapping cellular and molecular alterations hinder us from distinguishing the cause from the effect in AF pathogenesis. Yet, a plethora of therapeutic modalities target these molecular perturbations and hold promise in combating the AF burden. Namely, atrial selective ion channel inhibitors, AF gene therapy, anti-fibrotic agents, AF drug repurposing, immunomodulators, and indirect cardiac neuromodulation are discussed here.
Collapse
Affiliation(s)
- Konstantinos A. Papathanasiou
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | - Sotiria G. Giotaki
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | - Dimitrios A. Vrachatis
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | - Gerasimos Siasos
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | - Vaia Lambadiari
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | | | - Charalampos Kossyvakis
- Department of Cardiology, “G. Gennimatas” General Hospital of Athens, 11527 Athens, Greece; (C.K.); (A.K.); (K.R.); (G.D.); (D.A.)
| | - Andreas Kaoukis
- Department of Cardiology, “G. Gennimatas” General Hospital of Athens, 11527 Athens, Greece; (C.K.); (A.K.); (K.R.); (G.D.); (D.A.)
| | - Konstantinos Raisakis
- Department of Cardiology, “G. Gennimatas” General Hospital of Athens, 11527 Athens, Greece; (C.K.); (A.K.); (K.R.); (G.D.); (D.A.)
| | - Gerasimos Deftereos
- Department of Cardiology, “G. Gennimatas” General Hospital of Athens, 11527 Athens, Greece; (C.K.); (A.K.); (K.R.); (G.D.); (D.A.)
| | - Theodore G. Papaioannou
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | | | - Dimitrios Avramides
- Department of Cardiology, “G. Gennimatas” General Hospital of Athens, 11527 Athens, Greece; (C.K.); (A.K.); (K.R.); (G.D.); (D.A.)
| | - Spyridon G. Deftereos
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
- Correspondence: ; Tel.: +30-21-0583-2355
| |
Collapse
|
24
|
Fakuade FE, Tomsits P, Voigt N. Connexin hemichannels in atrial fibrillation: orphaned and irrelevant? Cardiovasc Res 2021; 117:4-6. [PMID: 33112373 DOI: 10.1093/cvr/cvaa308] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Funsho E Fakuade
- Institute of Pharmacology and Toxicology, University Medical Center, Robert-Koch-Str. 40, D-37075 Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany
| | - Philipp Tomsits
- Department of Medicine I, University Hospital Munich, Campus Großhadern, Ludwig-Maximilians University (LMU), Marchioninistraße 15, 81377 Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Germany.,Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians University (LMU), Marchioninistraße 27, 81377 Munich, Germany
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center, Robert-Koch-Str. 40, D-37075 Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany
| |
Collapse
|
25
|
Kany S, Reissmann B, Metzner A, Kirchhof P, Darbar D, Schnabel RB. Genetics of atrial fibrillation-practical applications for clinical management: if not now, when and how? Cardiovasc Res 2021; 117:1718-1731. [PMID: 33982075 PMCID: PMC8208749 DOI: 10.1093/cvr/cvab153] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
The prevalence and economic burden of atrial fibrillation (AF) are predicted to more than double over the next few decades. In addition to anticoagulation and treatment of concomitant cardiovascular conditions, early and standardized rhythm control therapy reduces cardiovascular outcomes as compared with a rate control approach, favouring the restoration, and maintenance of sinus rhythm safely. Current therapies for rhythm control of AF include antiarrhythmic drugs (AADs) and catheter ablation (CA). However, response in an individual patient is highly variable with some remaining free of AF for long periods on antiarrhythmic therapy, while others require repeat AF ablation within weeks. The limited success of rhythm control therapy for AF is in part related to incomplete understanding of the pathophysiological mechanisms and our inability to predict responses in individual patients. Thus, a major knowledge gap is predicting which patients with AF are likely to respond to rhythm control approach. Over the last decade, tremendous progress has been made in defining the genetic architecture of AF with the identification of rare mutations in cardiac ion channels, signalling molecules, and myocardial structural proteins associated with familial (early-onset) AF. Conversely, genome-wide association studies have identified common variants at over 100 genetic loci and the development of polygenic risk scores has identified high-risk individuals. Although retrospective studies suggest that response to AADs and CA is modulated in part by common genetic variation, the development of a comprehensive clinical and genetic risk score may enable the translation of genetic data to the bedside care of AF patients. Given the economic impact of the AF epidemic, even small changes in therapeutic efficacy may lead to substantial improvements for patients and health care systems.
Collapse
Affiliation(s)
- Shinwan Kany
- Department of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20251 Hamburg, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Martinistraße 52, 20251 Hamburg, Hamburg, Germany
| | - Bruno Reissmann
- Department of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20251 Hamburg, Hamburg, Germany
| | - Andreas Metzner
- Department of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20251 Hamburg, Hamburg, Germany
| | - Paulus Kirchhof
- Department of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20251 Hamburg, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Martinistraße 52, 20251 Hamburg, Hamburg, Germany.,The Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston Birmingham B15 2TT, UK
| | - Dawood Darbar
- Division of Cardiology, Departments of Medicine, University of Illinois at Chicago and Jesse Brown Veterans Administration, 840 South Wood Street, Suite 928 M/C 715, Chicago, IL 60612, USA
| | - Renate B Schnabel
- Department of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20251 Hamburg, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Martinistraße 52, 20251 Hamburg, Hamburg, Germany
| |
Collapse
|
26
|
De Smet MA, Lissoni A, Nezlobinsky T, Wang N, Dries E, Pérez-Hernández M, Lin X, Amoni M, Vervliet T, Witschas K, Rothenberg E, Bultynck G, Schulz R, Panfilov AV, Delmar M, Sipido KR, Leybaert L. Cx43 hemichannel microdomain signaling at the intercalated disc enhances cardiac excitability. J Clin Invest 2021; 131:137752. [PMID: 33621213 DOI: 10.1172/jci137752] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 02/19/2021] [Indexed: 12/11/2022] Open
Abstract
Cx43, a major cardiac connexin, forms precursor hemichannels that accrue at the intercalated disc to assemble as gap junctions. While gap junctions are crucial for electrical conduction in the heart, little is known about the potential roles of hemichannels. Recent evidence suggests that inhibiting Cx43 hemichannel opening with Gap19 has antiarrhythmic effects. Here, we used multiple electrophysiology, imaging, and super-resolution techniques to understand and define the conditions underlying Cx43 hemichannel activation in ventricular cardiomyocytes, their contribution to diastolic Ca2+ release from the sarcoplasmic reticulum, and their impact on electrical stability. We showed that Cx43 hemichannels were activated during diastolic Ca2+ release in single ventricular cardiomyocytes and cardiomyocyte cell pairs from mice and pigs. This activation involved Cx43 hemichannel Ca2+ entry and coupling to Ca2+ release microdomains at the intercalated disc, resulting in enhanced Ca2+ dynamics. Hemichannel opening furthermore contributed to delayed afterdepolarizations and triggered action potentials. In single cardiomyocytes, cardiomyocyte cell pairs, and arterially perfused tissue wedges from failing human hearts, increased hemichannel activity contributed to electrical instability compared with nonfailing rejected donor hearts. We conclude that microdomain coupling between Cx43 hemichannels and Ca2+ release is a potentially novel, targetable mechanism of cardiac arrhythmogenesis in heart failure.
Collapse
Affiliation(s)
- Maarten Aj De Smet
- Physiology Group, Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium.,Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium.,Department of Internal Medicine and
| | - Alessio Lissoni
- Physiology Group, Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Timur Nezlobinsky
- Department of Physics and Astronomy, Ghent University, Ghent, Belgium.,Laboratory of Computational Biology and Medicine, Ural Federal University, Ekaterinburg, Russia
| | - Nan Wang
- Physiology Group, Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Eef Dries
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Marta Pérez-Hernández
- Leon H. Charney Division of Cardiology, School of Medicine, New York University, New York, New York, USA
| | - Xianming Lin
- Leon H. Charney Division of Cardiology, School of Medicine, New York University, New York, New York, USA
| | - Matthew Amoni
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Tim Vervliet
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Katja Witschas
- Physiology Group, Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Eli Rothenberg
- Department of Biochemistry and Molecular Pharmacology, School of Medicine, New York University, New York, New York, USA
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Rainer Schulz
- Physiologisches Institut, Justus-Liebig-Universität, Giessen, Germany
| | - Alexander V Panfilov
- Department of Physics and Astronomy, Ghent University, Ghent, Belgium.,Laboratory of Computational Biology and Medicine, Ural Federal University, Ekaterinburg, Russia.,Arrhythmia Department, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Mario Delmar
- Leon H. Charney Division of Cardiology, School of Medicine, New York University, New York, New York, USA
| | - Karin R Sipido
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Luc Leybaert
- Physiology Group, Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
27
|
KLF15 Loss-of-Function Mutation Underlying Atrial Fibrillation as well as Ventricular Arrhythmias and Cardiomyopathy. Genes (Basel) 2021; 12:genes12030408. [PMID: 33809104 PMCID: PMC8001991 DOI: 10.3390/genes12030408] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/27/2021] [Accepted: 03/09/2021] [Indexed: 12/04/2022] Open
Abstract
Atrial fibrillation (AF) represents the most common type of clinical cardiac arrhythmia and substantially increases the risks of cerebral stroke, heart failure and death. Accumulating evidence has convincingly demonstrated the strong genetic basis of AF, and an increasing number of pathogenic variations in over 50 genes have been causally linked to AF. Nevertheless, AF is of pronounced genetic heterogeneity, and the genetic determinants underpinning AF in most patients remain obscure. In the current investigation, a Chinese pedigree with AF as well as ventricular arrhythmias and hypertrophic cardiomyopathy was recruited. Whole exome sequencing and bioinformatic analysis of the available family members were conducted, and a novel heterozygous variation in the KLF15 gene (encoding Krüppel-like factor 15, a transcription factor critical for cardiac electrophysiology and structural remodeling), NM_014079.4: c.685A>T; p.(Lys229*), was identified. The variation was verified by Sanger sequencing and segregated with autosomal dominant AF in the family with complete penetrance. The variation was absent from 300 unrelated healthy subjects used as controls. In functional assays using a dual-luciferase assay system, mutant KLF15 showed neither transcriptional activation of the KChIP2 promoter nor transcriptional inhibition of the CTGF promoter, alone or in the presence of TGFB1, a key player in the pathogenesis of arrhythmias and cardiomyopathies. The findings indicate KLF15 as a new causative gene responsible for AF as well as ventricular arrhythmias and hypertrophic cardiomyopathy, and they provide novel insight into the molecular mechanisms underlying cardiac arrhythmias and hypertrophic cardiomyopathy.
Collapse
|
28
|
The role of connexin proteins and their channels in radiation-induced atherosclerosis. Cell Mol Life Sci 2021; 78:3087-3103. [PMID: 33388835 PMCID: PMC8038956 DOI: 10.1007/s00018-020-03716-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/29/2020] [Accepted: 11/17/2020] [Indexed: 02/08/2023]
Abstract
Radiotherapy is an effective treatment for breast cancer and other thoracic tumors. However, while high-energy radiotherapy treatment successfully kills cancer cells, radiation exposure of the heart and large arteries cannot always be avoided, resulting in secondary cardiovascular disease in cancer survivors. Radiation-induced changes in the cardiac vasculature may thereby lead to coronary artery atherosclerosis, which is a major cardiovascular complication nowadays in thoracic radiotherapy-treated patients. The underlying biological and molecular mechanisms of radiation-induced atherosclerosis are complex and still not fully understood, resulting in potentially improper radiation protection. Ionizing radiation (IR) exposure may damage the vascular endothelium by inducing DNA damage, oxidative stress, premature cellular senescence, cell death and inflammation, which act to promote the atherosclerotic process. Intercellular communication mediated by connexin (Cx)-based gap junctions and hemichannels may modulate IR-induced responses and thereby the atherosclerotic process. However, the role of endothelial Cxs and their channels in atherosclerotic development after IR exposure is still poorly defined. A better understanding of the underlying biological pathways involved in secondary cardiovascular toxicity after radiotherapy would facilitate the development of effective strategies that prevent or mitigate these adverse effects. Here, we review the possible roles of intercellular Cx driven signaling and communication in radiation-induced atherosclerosis.
Collapse
|
29
|
Samuel RM, Majd H, Richter MN, Ghazizadeh Z, Zekavat SM, Navickas A, Ramirez JT, Asgharian H, Simoneau CR, Bonser LR, Koh KD, Garcia-Knight M, Tassetto M, Sunshine S, Farahvashi S, Kalantari A, Liu W, Andino R, Zhao H, Natarajan P, Erle DJ, Ott M, Goodarzi H, Fattahi F. Androgen Signaling Regulates SARS-CoV-2 Receptor Levels and Is Associated with Severe COVID-19 Symptoms in Men. Cell Stem Cell 2020; 27:876-889.e12. [PMID: 33232663 PMCID: PMC7670929 DOI: 10.1016/j.stem.2020.11.009] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 10/17/2020] [Accepted: 11/13/2020] [Indexed: 01/08/2023]
Abstract
SARS-CoV-2 infection has led to a global health crisis, and yet our understanding of the disease and potential treatment options remains limited. The infection occurs through binding of the virus with angiotensin converting enzyme 2 (ACE2) on the cell membrane. Here, we established a screening strategy to identify drugs that reduce ACE2 levels in human embryonic stem cell (hESC)-derived cardiac cells and lung organoids. Target analysis of hit compounds revealed androgen signaling as a key modulator of ACE2 levels. Treatment with antiandrogenic drugs reduced ACE2 expression and protected hESC-derived lung organoids against SARS-CoV-2 infection. Finally, clinical data on COVID-19 patients demonstrated that prostate diseases, which are linked to elevated androgen, are significant risk factors and that genetic variants that increase androgen levels are associated with higher disease severity. These findings offer insights on the mechanism of disproportionate disease susceptibility in men and identify antiandrogenic drugs as candidate therapeutics for COVID-19.
Collapse
Affiliation(s)
- Ryan M Samuel
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
| | - Homa Majd
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
| | - Mikayla N Richter
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
| | - Zaniar Ghazizadeh
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Yale School of Medicine, New Haven, CT 06510, USA
| | - Seyedeh Maryam Zekavat
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Program of Computational Biology & Bioinformatics, Yale University, New Haven, CT 06510, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Albertas Navickas
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Jonathan T Ramirez
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
| | - Hosseinali Asgharian
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | | | - Luke R Bonser
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kyung Duk Koh
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Miguel Garcia-Knight
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michel Tassetto
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sara Sunshine
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Sina Farahvashi
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
| | - Ali Kalantari
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
| | - Wei Liu
- Program of Computational Biology & Bioinformatics, Yale University, New Haven, CT 06510, USA
| | - Raul Andino
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hongyu Zhao
- Program of Computational Biology & Bioinformatics, Yale University, New Haven, CT 06510, USA; Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Pradeep Natarajan
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - David J Erle
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Melanie Ott
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Hani Goodarzi
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA; Department of Urology, University of California, San Francisco, CA 94158, USA; Bakar Computational Health Sciences Institute, University of California, San Francisco, CA 94158, USA.
| | - Faranak Fattahi
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA; Program in Craniofacial Biology, University of California, San Francisco, CA 94110, USA.
| |
Collapse
|
30
|
Lissoni A, Wang N, Nezlobinskii T, De Smet M, Panfilov AV, Vandersickel N, Leybaert L, Witschas K. Gap19, a Cx43 Hemichannel Inhibitor, Acts as a Gating Modifier That Decreases Main State Opening While Increasing Substate Gating. Int J Mol Sci 2020; 21:ijms21197340. [PMID: 33027889 PMCID: PMC7583728 DOI: 10.3390/ijms21197340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/12/2020] [Accepted: 09/25/2020] [Indexed: 02/05/2023] Open
Abstract
Cx43 hemichannels (HCs) are electrically and chemically gated transmembrane pores with low open probability and multiple conductance states, which makes kinetic studies of channel gating in large datasets challenging. Here, we developed open access software, named HemiGUI, to analyze HC gating transitions and investigated voltage-induced HC opening based on up to ≈4000 events recorded in HeLa-Cx43-overexpressing cells. We performed a detailed characterization of Cx43 HC gating profiles and specifically focused on the role of the C-terminal tail (CT) domain by recording the impact of adding an EGFP tag to the Cx43 CT end (Cx43-EGFP) or by supplying the Cx43 HC-inhibiting peptide Gap19 that interferes with CT interaction with the cytoplasmic loop (CL). We found that Gap19 not only decreased HC opening activity to the open state (≈217 pS) but also increased the propensity of subconductance (≈80 pS) transitions that additionally became slower as compared to the control. The work demonstrates that large sample transition analysis allows detailed investigations on Cx43 HC gating and shows that Gap19 acts as a HC gating modifier by interacting with the CT that forms a crucial gating element.
Collapse
Affiliation(s)
- Alessio Lissoni
- Department of Basic and Applied Medical Sciences—Physiology Group, Ghent University, 9000 Ghent, Belgium; (A.L.); (N.W.); (M.D.S.)
| | - Nan Wang
- Department of Basic and Applied Medical Sciences—Physiology Group, Ghent University, 9000 Ghent, Belgium; (A.L.); (N.W.); (M.D.S.)
| | - Timur Nezlobinskii
- Department of Physics and Astronomy, Ghent University, 9000 Ghent, Belgium; (T.N.); (A.V.P.); (N.V.)
| | - Maarten De Smet
- Department of Basic and Applied Medical Sciences—Physiology Group, Ghent University, 9000 Ghent, Belgium; (A.L.); (N.W.); (M.D.S.)
| | - Alexander V. Panfilov
- Department of Physics and Astronomy, Ghent University, 9000 Ghent, Belgium; (T.N.); (A.V.P.); (N.V.)
- Laboratory of Computational Biology and Medicine, Ural Federal University, 620075 Ekaterinburg, Russia
| | - Nele Vandersickel
- Department of Physics and Astronomy, Ghent University, 9000 Ghent, Belgium; (T.N.); (A.V.P.); (N.V.)
| | - Luc Leybaert
- Department of Basic and Applied Medical Sciences—Physiology Group, Ghent University, 9000 Ghent, Belgium; (A.L.); (N.W.); (M.D.S.)
- Correspondence: (L.L.); (K.W.); Tel.: +32-9-332-3366 (L.L.); +32-9-332-6944 (K.W.)
| | - Katja Witschas
- Department of Basic and Applied Medical Sciences—Physiology Group, Ghent University, 9000 Ghent, Belgium; (A.L.); (N.W.); (M.D.S.)
- Correspondence: (L.L.); (K.W.); Tel.: +32-9-332-3366 (L.L.); +32-9-332-6944 (K.W.)
| |
Collapse
|
31
|
Bektik E, Cowan DB, Wang DZ. Long Non-Coding RNAs in Atrial Fibrillation: Pluripotent Stem Cell-Derived Cardiomyocytes as a Model System. Int J Mol Sci 2020; 21:ijms21155424. [PMID: 32751460 PMCID: PMC7432754 DOI: 10.3390/ijms21155424] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is a type of sustained arrhythmia in humans often characterized by devastating alterations to the cardiac conduction system as well as the structure of the atria. AF can lead to decreased cardiac function, heart failure, and other complications. Long non-coding RNAs (lncRNAs) have been shown to play important roles in the cardiovascular system, including AF; however, a large group of lncRNAs is not conserved between mouse and human. Furthermore, AF has complex networks showing variations in mechanisms in different species, making it challenging to utilize conventional animal models to investigate the functional roles and potential therapeutic benefits of lncRNAs for AF. Fortunately, pluripotent stem cell (PSC)-derived cardiomyocytes (CMs) offer a reliable platform to study lncRNA functions in AF because of certain electrophysiological and molecular similarities with native human CMs. In this review, we first summarize the broad aspects of lncRNAs in various heart disease settings, then focus on their potential roles in AF development and pathophysiology. We also discuss current uses of PSCs in AF research and describe how these studies could be developed into novel therapeutics for AF and other cardiovascular diseases.
Collapse
Affiliation(s)
- Emre Bektik
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood, Boston, MA 02115, USA; (E.B.); (D.B.C.)
| | - Douglas B. Cowan
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood, Boston, MA 02115, USA; (E.B.); (D.B.C.)
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood, Boston, MA 02115, USA; (E.B.); (D.B.C.)
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Correspondence:
| |
Collapse
|
32
|
Ghazizadeh Z, Majd H, Richter M, Samuel R, Zekavat SM, Asgharian H, Farahvashi S, Kalantari A, Ramirez J, Zhao H, Natarajan P, Goodarzi H, Fattahi F. Androgen Regulates SARS-CoV-2 Receptor Levels and Is Associated with Severe COVID-19 Symptoms in Men. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.05.12.091082. [PMID: 32511360 PMCID: PMC7263488 DOI: 10.1101/2020.05.12.091082] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has led to a global health crisis, and yet our understanding of the disease pathophysiology and potential treatment options remains limited. SARS-CoV-2 infection occurs through binding and internalization of the viral spike protein to angiotensin converting enzyme 2 (ACE2) on the host cell membrane. Lethal complications are caused by damage and failure of vital organs that express high levels of ACE2, including the lungs, the heart and the kidneys. Here, we established a high-throughput drug screening strategy to identify therapeutic candidates that reduce ACE2 levels in human embryonic stem cell (hESC) derived cardiac cells. Drug target analysis of validated hit compounds, including 5 alpha reductase inhibitors, revealed androgen signaling as a key modulator of ACE2 levels. Treatment with the 5 alpha reductase inhibitor dutasteride reduced ACE2 levels and internalization of recombinant spike receptor binding domain (Spike-RBD) in hESC-derived cardiac cells and human alveolar epithelial cells. Finally, clinical data on coronavirus disease 2019 (COVID-19) patients demonstrated that abnormal androgen states are significantly associated with severe disease complications and cardiac injury as measured by blood troponin T levels. These findings provide important insights on the mechanism of increased disease susceptibility in male COVID-19 patients and identify androgen receptor inhibition as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Zaniar Ghazizadeh
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06510
- Yale School of Medicine, New Haven, CT, 06510
| | - Homa Majd
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143
| | - Mikayla Richter
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143
| | - Ryan Samuel
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143
| | - Seyedeh Maryam Zekavat
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142
- Department of computational Biology & Bioinformatics, Yale University, New Haven, CT, 06510
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114
| | - Hosseinali Asgharian
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158
| | - Sina Farahvashi
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143
| | - Ali Kalantari
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143
| | - Jonathan Ramirez
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143
| | - Hongyu Zhao
- Department of computational Biology & Bioinformatics, Yale University, New Haven, CT, 06510
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, 06510
| | - Pradeep Natarajan
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114
- Department of Medicine, Harvard Medical School, Boston, MA 02115
| | - Hani Goodarzi
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158
- Department of Urology, University of California, San Francisco, CA 94158
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA 94158
| | - Faranak Fattahi
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158
- Program in Craniofacial Biology, University of California, San Francisco, CA 94110 USA
| |
Collapse
|