1
|
An N, Yang F, Zhang G, Jiang Y, Liu H, Gao Y, Li Y, Ji P, Shang H, Xing Y. Single-cell RNA sequencing reveals the contribution of smooth muscle cells and endothelial cells to fibrosis in human atrial tissue with atrial fibrillation. Mol Med 2024; 30:247. [PMID: 39701940 DOI: 10.1186/s10020-024-00999-1] [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: 09/30/2024] [Accepted: 11/13/2024] [Indexed: 12/21/2024] Open
Abstract
AIMS Atrial fibrillation (AF) has high mortality and morbidity rates. However, the intracellular molecular complexity of the atrial tissue of patients with AF has not been adequately assessed. METHODS AND RESULTS We investigated the cellular heterogeneity of human atrial tissue and changes in differentially expressed genes between cells using single-cell RNA sequencing, fluorescence in situ hybridization, intercellular communication, and cell trajectory analysis. Using genome-wide association studies (GWAS) and proteomics, we discovered cell types enriched for AF susceptibility genes. We discovered eight different cell types, which were further subdivided into 23 subpopulations. In AF, the communication strength between smooth muscle cells (SMCs) and fibroblast (FB) 3 cells increased and the relevant signaling pathways were quite similar. Subpopulations of endothelial cells (ECs) are mainly involved in fibrosis through TXNDC5 and POSTN. AF susceptibility genes revealed by GWAS were especially enriched in neuronal and epicardial cells, FB3, and lymphoid (Lys) cells, whereas proteomic sequencing differential proteins were concentrated in FB3 cells and SMCs. CONCLUSIONS This study provides a cellular landscape based on the atrial tissue of patients with AF and highlights intercellular changes and differentially expressed genes that occur during the disease process. A thorough description of the cellular populations involved in AF will facilitate the identification of new cell-based interventional targets with direct functional significance for the treatment of human disease.
Collapse
Affiliation(s)
- Na An
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, No.5 Beixian'ge Street, Xicheng District, Beijing, 100053, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, No.5 Haiyuncang, Dongcheng District, Beijing, 100700, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Fan Yang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, No.5 Beixian'ge Street, Xicheng District, Beijing, 100053, China
| | - Guoxia Zhang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, No.5 Beixian'ge Street, Xicheng District, Beijing, 100053, China
| | - Yuchen Jiang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, No.5 Beixian'ge Street, Xicheng District, Beijing, 100053, China
| | - Haoqi Liu
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, No.5 Haiyuncang, Dongcheng District, Beijing, 100700, China
| | - Yingjian Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Peifeng Ji
- Computational Genomics Lab, Beijing Institutes of Life Science, Chinese Academy of Sciences, No. 5, Yard 1, Beichen West Road, Chaoyang District, Beijing, 100101, China.
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, No.5 Haiyuncang, Dongcheng District, Beijing, 100700, China.
| | - Yanwei Xing
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, No.5 Beixian'ge Street, Xicheng District, Beijing, 100053, China.
| |
Collapse
|
2
|
Paucar M, Nilsson D, Engvall M, Laffita-Mesa J, Söderhäll C, Skorpil M, Halldin C, Fazio P, Lagerstedt-Robinson K, Solders G, Angeria M, Varrone A, Risling M, Jiao H, Nennesmo I, Wedell A, Svenningsson P. Spinocerebellar ataxia type 4 is caused by a GGC expansion in the ZFHX3 gene and is associated with prominent dysautonomia and motor neuron signs. J Intern Med 2024; 296:234-248. [PMID: 38973251 DOI: 10.1111/joim.13815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
BACKGROUND Spinocerebellar ataxia 4 (SCA4), characterized in 1996, features adult-onset ataxia, polyneuropathy, and linkage to chromosome 16q22.1; its underlying mutation has remained elusive. OBJECTIVE To explore the radiological and neuropathological abnormalities in the entire neuroaxis in SCA4 and search for its mutation. METHODS Three Swedish families with undiagnosed ataxia went through clinical, neurophysiological, and neuroimaging tests, including PET studies and genetic investigations. In four cases, neuropathological assessments of the neuroaxis were performed. Genetic testing included short read whole genome sequencing, short tandem repeat analysis with ExpansionHunter de novo, and long read sequencing. RESULTS Novel features for SCA4 include dysautonomia, motor neuron affection, and abnormal eye movements. We found evidence of anticipation; neuroimaging demonstrated atrophy in the cerebellum, brainstem, and spinal cord. [18F]FDG-PET demonstrated brain hypometabolism and [11C]Flumazenil-PET reduced binding in several brain lobes, insula, thalamus, hypothalamus, and cerebellum. Moderate to severe loss of Purkinje cells in the cerebellum and of motor neurons in the anterior horns of the spinal cord along with pronounced degeneration of posterior tracts was also found. Intranuclear, mainly neuronal, inclusions positive for p62 and ubiquitin were sparse but widespread in the CNS. This finding prompted assessment for nucleotide expansions. A polyglycine stretch encoding GGC expansions in the last exon of the zink finger homeobox 3 gene was identified segregating with disease and not found in 1000 controls. CONCLUSIONS SCA4 is a neurodegenerative disease caused by a novel GGC expansion in the coding region of ZFHX3, and its spectrum is expanded to include dysautonomia and neuromuscular manifestations.
Collapse
Affiliation(s)
- Martin Paucar
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Daniel Nilsson
- Department of Clinical Genetics and Genomics, Karolinska University Hospital, Stockholm, Sweden
- Science for Life Laboratory, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Martin Engvall
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - José Laffita-Mesa
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Cilla Söderhäll
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Skorpil
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Christer Halldin
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Patrik Fazio
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Kristina Lagerstedt-Robinson
- Department of Clinical Genetics and Genomics, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Göran Solders
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurophysiology, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Angeria
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Andrea Varrone
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Mårten Risling
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Hong Jiao
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Inger Nennesmo
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Anna Wedell
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Per Svenningsson
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
3
|
Petzl AM, Jabbour G, Cadrin-Tourigny J, Pürerfellner H, Macle L, Khairy P, Avram R, Tadros R. Innovative approaches to atrial fibrillation prediction: should polygenic scores and machine learning be implemented in clinical practice? Europace 2024; 26:euae201. [PMID: 39073570 PMCID: PMC11332604 DOI: 10.1093/europace/euae201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Atrial fibrillation (AF) prediction and screening are of important clinical interest because of the potential to prevent serious adverse events. Devices capable of detecting short episodes of arrhythmia are now widely available. Although it has recently been suggested that some high-risk patients with AF detected on implantable devices may benefit from anticoagulation, long-term management remains challenging in lower-risk patients and in those with AF detected on monitors or wearable devices as the development of clinically meaningful arrhythmia burden in this group remains unknown. Identification and prediction of clinically relevant AF is therefore of unprecedented importance to the cardiologic community. Family history and underlying genetic markers are important risk factors for AF. Recent studies suggest a good predictive ability of polygenic risk scores, with a possible additive value to clinical AF prediction scores. Artificial intelligence, enabled by the exponentially increasing computing power and digital data sets, has gained traction in the past decade and is of increasing interest in AF prediction using a single or multiple lead sinus rhythm electrocardiogram. Integrating these novel approaches could help predict AF substrate severity, thereby potentially improving the effectiveness of AF screening and personalizing the management of patients presenting with conditions such as embolic stroke of undetermined source or subclinical AF. This review presents current evidence surrounding deep learning and polygenic risk scores in the prediction of incident AF and provides a futuristic outlook on possible ways of implementing these modalities into clinical practice, while considering current limitations and required areas of improvement.
Collapse
Affiliation(s)
- Adrian M Petzl
- Electrophysiology Service, Department of Medicine, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
- Cardiovascular Genetics Center, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
| | - Gilbert Jabbour
- Electrophysiology Service, Department of Medicine, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
- Heartwise (heartwise.ai), Montreal Heart Institute, Montreal, Canada
| | - Julia Cadrin-Tourigny
- Electrophysiology Service, Department of Medicine, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
- Cardiovascular Genetics Center, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
| | - Helmut Pürerfellner
- Department of Internal Medicine 2/Cardiology, Ordensklinikum Linz Elisabethinen, Linz, Austria
| | - Laurent Macle
- Electrophysiology Service, Department of Medicine, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
| | - Paul Khairy
- Electrophysiology Service, Department of Medicine, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
| | - Robert Avram
- Heartwise (heartwise.ai), Montreal Heart Institute, Montreal, Canada
- Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Rafik Tadros
- Electrophysiology Service, Department of Medicine, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
- Cardiovascular Genetics Center, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
| |
Collapse
|
4
|
Owais A, Barney M, Ly OT, Brown G, Chen H, Sridhar A, Pavel A, Khetani SR, Darbar D. Genetics and Pharmacogenetics of Atrial Fibrillation: A Mechanistic Perspective. JACC Basic Transl Sci 2024; 9:918-934. [PMID: 39170958 PMCID: PMC11334418 DOI: 10.1016/j.jacbts.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 08/23/2024]
Abstract
The heritability of atrial fibrillation (AF) is well established. Over the last decade genetic architecture of AF has been unraveled by genome-wide association studies and family-based studies. However, the translation of these genetic discoveries has lagged owing to an incomplete understanding of the pathogenic mechanisms underlying the genetic variants, challenges in classifying variants of uncertain significance (VUS), and limitations of existing disease models. We review the mechanistic insight provided by basic science studies regarding AF mechanisms, recent developments in high-throughput classification of VUS, and advances in bioengineered cardiac models for developing personalized therapy for AF.
Collapse
Affiliation(s)
- Asia Owais
- Division of Cardiology, Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Miles Barney
- Division of Cardiology, Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Olivia Thao Ly
- Division of Cardiology, Department of Medicine, University of Illinois, Chicago, Illinois, USA
- Department of Biomedical Engineering, University of Illinois, Chicago, Illinois, USA
| | - Grace Brown
- Division of Cardiology, Department of Medicine, University of Illinois, Chicago, Illinois, USA
- Department of Biomedical Engineering, University of Illinois, Chicago, Illinois, USA
| | - Hanna Chen
- Division of Cardiology, Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Arvind Sridhar
- Division of Cardiology, Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Arif Pavel
- Division of Cardiology, Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Salman R. Khetani
- Department of Biomedical Engineering, University of Illinois, Chicago, Illinois, USA
| | - Dawood Darbar
- Division of Cardiology, Department of Medicine, University of Illinois, Chicago, Illinois, USA
- Department of Biomedical Engineering, University of Illinois, Chicago, Illinois, USA
- Department of Physiology and Biophysics, University of Illinois, Chicago, Illinois, USA
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago, Illinois, USA
| |
Collapse
|
5
|
Kukendrarajah K, Farmaki AE, Lambiase PD, Schilling R, Finan C, Floriaan Schmidt A, Providencia R. Advancing drug development for atrial fibrillation by prioritising findings from human genetic association studies. EBioMedicine 2024; 105:105194. [PMID: 38941956 PMCID: PMC11260865 DOI: 10.1016/j.ebiom.2024.105194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 05/14/2024] [Accepted: 05/28/2024] [Indexed: 06/30/2024] Open
Abstract
BACKGROUND Drug development for atrial fibrillation (AF) has failed to yield new approved compounds. We sought to identify and prioritise potential druggable targets with support from human genetics, by integrating the available evidence with bioinformatics sources relevant for AF drug development. METHODS Genetic hits for AF and related traits were identified through structured search of MEDLINE. Genes derived from each paper were cross-referenced with the OpenTargets platform for drug interactions. Confirmation/validation was demonstrated through structured searches and review of evidence on MEDLINE and ClinialTrials.gov for each drug and its association with AF. FINDINGS 613 unique drugs were identified, with 21 already included in AF Guidelines. Cardiovascular drugs from classes not currently used for AF (e.g. ranolazine and carperitide) and anti-inflammatory drugs (e.g. dexamethasone and mehylprednisolone) had evidence of potential benefit. Further targets were considered druggable but remain open for drug development. INTERPRETATION Our systematic approach, combining evidence from different bioinformatics platforms, identified drug repurposing opportunities and druggable targets for AF. FUNDING KK is supported by Barts Charity grant G-002089 and is mentored on the AFGen 2023-24 Fellowship funded by the AFGen NIH/NHLBI grant R01HL092577. RP is supported by the UCL BHF Research Accelerator AA/18/6/34223 and NIHR grant NIHR129463. AFS is supported by the BHF grants PG/18/5033837, PG/22/10989 and UCL BHF Accelerator AA/18/6/34223 as well as the UK Research and Innovation (UKRI) under the UK government's Horizon Europe funding guarantee EP/Z000211/1 and by the UKRI-NIHR grant MR/V033867/1 for the Multimorbidity Mechanism and Therapeutics Research Collaboration. AF is supported by UCL BHF Accelerator AA/18/6/34223. CF is supported by UCL BHF Accelerator AA/18/6/34223.
Collapse
Affiliation(s)
- Kishore Kukendrarajah
- Institute of Health Informatics, University College London, 222 Euston Road, NW1 2DA, United Kingdom; Barts Heart Centre, St Bartholomew's Hospital, West Smithfield, EC1A 7BE, United Kingdom.
| | - Aliki-Eleni Farmaki
- Institute of Health Informatics, University College London, 222 Euston Road, NW1 2DA, United Kingdom
| | - Pier D Lambiase
- Barts Heart Centre, St Bartholomew's Hospital, West Smithfield, EC1A 7BE, United Kingdom; Institute of Cardiovascular Science, University College London, Gower Street, WC1E 6HX, United Kingdom
| | - Richard Schilling
- Barts Heart Centre, St Bartholomew's Hospital, West Smithfield, EC1A 7BE, United Kingdom
| | - Chris Finan
- Institute of Cardiovascular Science, University College London, Gower Street, WC1E 6HX, United Kingdom; UCL British Heart Foundation Research Accelerator, United Kingdom; Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Amand Floriaan Schmidt
- Institute of Cardiovascular Science, University College London, Gower Street, WC1E 6HX, United Kingdom; UCL British Heart Foundation Research Accelerator, United Kingdom; Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands; Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centres, University of Amsterdam, the Netherlands
| | - Rui Providencia
- Institute of Health Informatics, University College London, 222 Euston Road, NW1 2DA, United Kingdom; Barts Heart Centre, St Bartholomew's Hospital, West Smithfield, EC1A 7BE, United Kingdom
| |
Collapse
|
6
|
Pérez Baca MDR, Jacobs EZ, Vantomme L, Leblanc P, Bogaert E, Dheedene A, De Cock L, Haghshenas S, Foroutan A, Levy MA, Kerkhof J, McConkey H, Chen CA, Batzir NA, Wang X, Palomares M, Carels M, Dermaut B, Sadikovic B, Menten B, Yuan B, Vergult S, Callewaert B. Haploinsufficiency of ZFHX3, encoding a key player in neuronal development, causes syndromic intellectual disability. Am J Hum Genet 2024; 111:509-528. [PMID: 38412861 PMCID: PMC10940049 DOI: 10.1016/j.ajhg.2024.01.013] [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: 06/07/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/29/2024] Open
Abstract
Neurodevelopmental disorders (NDDs) result from impaired development and functioning of the brain. Here, we identify loss-of-function (LoF) variation in ZFHX3 as a cause for syndromic intellectual disability (ID). ZFHX3 is a zinc-finger homeodomain transcription factor involved in various biological processes, including cell differentiation and tumorigenesis. We describe 42 individuals with protein-truncating variants (PTVs) or (partial) deletions of ZFHX3, exhibiting variable intellectual disability and autism spectrum disorder, recurrent facial features, relative short stature, brachydactyly, and, rarely, cleft palate. ZFHX3 LoF associates with a specific methylation profile in whole blood extracted DNA. Nuclear abundance of ZFHX3 increases during human brain development and neuronal differentiation. ZFHX3 was found to interact with the chromatin remodeling BRG1/Brm-associated factor complex and the cleavage and polyadenylation complex, suggesting a function in chromatin remodeling and mRNA processing. Furthermore, ChIP-seq for ZFHX3 revealed that it predominantly binds promoters of genes involved in nervous system development. We conclude that loss-of-function variants in ZFHX3 are a cause of syndromic ID associating with a specific DNA methylation profile.
Collapse
Affiliation(s)
- María Del Rocío Pérez Baca
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Eva Z Jacobs
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Lies Vantomme
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Pontus Leblanc
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Elke Bogaert
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Annelies Dheedene
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Laurenz De Cock
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Sadegheh Haghshenas
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Aidin Foroutan
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 3K7, Canada; Children's Health Research Institute, Lawson Research Institute, London, ON N6C 2R5, Canada
| | - Michael A Levy
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Jennifer Kerkhof
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Haley McConkey
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada; Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 3K7, Canada
| | - Chun-An Chen
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - Nurit Assia Batzir
- Schneider Children's Medical Center of Israel, Petach Tikvah 4920235, Israel
| | - Xia Wang
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - María Palomares
- INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario la Paz, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - Marieke Carels
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; VIB UGent Center for Inflammation Research, Department for Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Bart Dermaut
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Bekim Sadikovic
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada; Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 3K7, Canada
| | - Björn Menten
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Bo Yuan
- Seattle Children's Hospital, Seattle and Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98105, USA
| | - Sarah Vergult
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium.
| | - Bert Callewaert
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
7
|
Rudaks LI, Yeow D, Kumar KR. SCA4 Unravelled After More than 25 Years Using Advanced Genomic Technologies. Mov Disord 2024; 39:457-461. [PMID: 38525586 DOI: 10.1002/mds.29738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 03/26/2024] Open
Affiliation(s)
- Laura Ivete Rudaks
- Translational Neurogenomics Group, Concord Repatriation General Hospital, Concord, Australia
- Molecular Medicine Laboratory and Neurology Department, Concord Repatriation General Hospital, Concord, Australia
- Concord Clinical School, Faculty of Medicine and Health, The University of Sydney, Concord, Australia
- Genomic and Inherited Disease Program, The Garvan Institute of Medical Research, Darlinghurst, Australia
- Clinical Genetics Unit, Royal North Shore Hospital, St Leonards, Australia
| | - Dennis Yeow
- Translational Neurogenomics Group, Concord Repatriation General Hospital, Concord, Australia
- Molecular Medicine Laboratory and Neurology Department, Concord Repatriation General Hospital, Concord, Australia
- Concord Clinical School, Faculty of Medicine and Health, The University of Sydney, Concord, Australia
- Genomic and Inherited Disease Program, The Garvan Institute of Medical Research, Darlinghurst, Australia
- Neurodegenerative Service, Prince of Wales Hospital, Randwick, Australia
- Neuroscience Research Australia, Randwick, Australia
| | - Kishore Raj Kumar
- Translational Neurogenomics Group, Concord Repatriation General Hospital, Concord, Australia
- Molecular Medicine Laboratory and Neurology Department, Concord Repatriation General Hospital, Concord, Australia
- Concord Clinical School, Faculty of Medicine and Health, The University of Sydney, Concord, Australia
- Genomic and Inherited Disease Program, The Garvan Institute of Medical Research, Darlinghurst, Australia
- St Vincent's Healthcare Campus, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Darlinghurst, Australia
| |
Collapse
|
8
|
Chen Z, Gustavsson EK, Macpherson H, Anderson C, Clarkson C, Rocca C, Self E, Alvarez Jerez P, Scardamaglia A, Pellerin D, Montgomery K, Lee J, Gagliardi D, Luo H, Hardy J, Polke J, Singleton AB, Blauwendraat C, Mathews KD, Tucci A, Fu YH, Houlden H, Ryten M, Ptáček LJ. Adaptive Long-Read Sequencing Reveals GGC Repeat Expansion in ZFHX3 Associated with Spinocerebellar Ataxia Type 4. Mov Disord 2024; 39:486-497. [PMID: 38197134 DOI: 10.1002/mds.29704] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/29/2023] [Accepted: 12/15/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Spinocerebellar ataxia type 4 (SCA4) is an autosomal dominant ataxia with invariable sensory neuropathy originally described in a family with Swedish ancestry residing in Utah more than 25 years ago. Despite tight linkage to the 16q22 region, the molecular diagnosis has since remained elusive. OBJECTIVES Inspired by pathogenic structural variation implicated in other 16q-ataxias with linkage to the same locus, we revisited the index SCA4 cases from the Utah family using novel technologies to investigate structural variation within the candidate region. METHODS We adopted a targeted long-read sequencing approach with adaptive sampling on the Oxford Nanopore Technologies (ONT) platform that enables the detection of segregating structural variants within a genomic region without a priori assumptions about any variant features. RESULTS Using this approach, we found a heterozygous (GGC)n repeat expansion in the last coding exon of the zinc finger homeobox 3 (ZFHX3) gene that segregates with disease, ranging between 48 and 57 GGC repeats in affected probands. This finding was replicated in a separate family with SCA4. Furthermore, the estimation of this GGC repeat size in short-read whole genome sequencing (WGS) data of 21,836 individuals recruited to the 100,000 Genomes Project in the UK and our in-house dataset of 11,258 exomes did not reveal any pathogenic repeats, indicating that the variant is ultrarare. CONCLUSIONS These findings support the utility of adaptive long-read sequencing as a powerful tool to decipher causative structural variation in unsolved cases of inherited neurological disease. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Zhongbo Chen
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, United Kingdom
| | - Emil K Gustavsson
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, United Kingdom
| | - Hannah Macpherson
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Claire Anderson
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, United Kingdom
| | - Chris Clarkson
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Clarissa Rocca
- Department of Neuromuscular Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Eleanor Self
- Department of Neuromuscular Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Pilar Alvarez Jerez
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Annarita Scardamaglia
- Department of Neuromuscular Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - David Pellerin
- Department of Neuromuscular Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Kylie Montgomery
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, United Kingdom
| | - Jasmaine Lee
- Department of Neuromuscular Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Delia Gagliardi
- Department of Neuromuscular Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Huihui Luo
- Department of Neuromuscular Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - John Hardy
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
- Reta Lila Weston Institute, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UK Dementia Research Institute, University College London, London, United Kingdom
- NIHR University College London Hospitals Biomedical Research Centre, London, United Kingdom
- Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong, China
| | - James Polke
- The Neurogenetics Laboratory, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Andrew B Singleton
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Cornelis Blauwendraat
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Katherine D Mathews
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
- Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Arianna Tucci
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Ying-Hui Fu
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, USA
- Weill Institute for Neuroscience, University of California San Francisco, San Francisco, California, USA
- Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, California, USA
| | - Henry Houlden
- Department of Neuromuscular Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
- The Neurogenetics Laboratory, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Mina Ryten
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, United Kingdom
| | - Louis J Ptáček
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, USA
- Weill Institute for Neuroscience, University of California San Francisco, San Francisco, California, USA
- Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
9
|
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
|
10
|
Casalino-Matsuda SM, Chen F, Gonzalez-Gonzalez FJ, Matsuda H, Nair A, Abdala-Valencia H, Budinger GS, Dong JT, Beitel GJ, Sporn PH. Myeloid Zfhx3 deficiency protects against hypercapnia-induced suppression of host defense against influenza A virus. JCI Insight 2024; 9:e170316. [PMID: 38227369 PMCID: PMC11143927 DOI: 10.1172/jci.insight.170316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 01/10/2024] [Indexed: 01/17/2024] Open
Abstract
Hypercapnia, elevation of the partial pressure of CO2 in blood and tissues, is a risk factor for mortality in patients with severe acute and chronic lung diseases. We previously showed that hypercapnia inhibits multiple macrophage and neutrophil antimicrobial functions and that elevated CO2 increases the mortality of bacterial and viral pneumonia in mice. Here, we show that normoxic hypercapnia downregulates innate immune and antiviral gene programs in alveolar macrophages (AMØs). We also show that zinc finger homeobox 3 (Zfhx3) - a mammalian ortholog of zfh2, which mediates hypercapnic immune suppression in Drosophila - is expressed in mouse and human macrophages. Deletion of Zfhx3 in the myeloid lineage blocked the suppressive effect of hypercapnia on immune gene expression in AMØs and decreased viral replication, inflammatory lung injury, and mortality in hypercapnic mice infected with influenza A virus. To our knowledge, our results establish Zfhx3 as the first known mammalian mediator of CO2 effects on immune gene expression and lay the basis for future studies to identify therapeutic targets to interrupt hypercapnic immunosuppression in patients with advanced lung disease.
Collapse
Affiliation(s)
- S. Marina Casalino-Matsuda
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Fei Chen
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Francisco J. Gonzalez-Gonzalez
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Hiroaki Matsuda
- Department of Physical Sciences and Engineering, Wilbur Wright College, Chicago, Illinois, USA
| | - Aisha Nair
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Hiam Abdala-Valencia
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - G.R. Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Research Service, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Jin-Tang Dong
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Greg J. Beitel
- Department of Molecular Biosciences, Weinberg College of Arts and Sciences, Northwestern University, Evanston, Illinois, USA
| | - Peter H.S. Sporn
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Research Service, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| |
Collapse
|
11
|
Grzeczka A, Graczyk S, Kordowitzki P. DNA Methylation and Telomeres-Their Impact on the Occurrence of Atrial Fibrillation during Cardiac Aging. Int J Mol Sci 2023; 24:15699. [PMID: 37958686 PMCID: PMC10650750 DOI: 10.3390/ijms242115699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Atrial fibrillation (AF) is the most common arrhythmia in humans. AF is characterized by irregular and increased atrial muscle activation. This high-frequency activation obliterates the synchronous work of the atria and ventricles, reducing myocardial performance, which can lead to severe heart failure or stroke. The risk of developing atrial fibrillation depends largely on the patient's history. Cardiovascular diseases are considered aging-related pathologies; therefore, deciphering the role of telomeres and DNA methylation (mDNA), two hallmarks of aging, is likely to contribute to a better understanding and prophylaxis of AF. In honor of Prof. Elizabeth Blackburn's 75th birthday, we dedicate this review to the discovery of telomeres and her contribution to research on aging.
Collapse
Affiliation(s)
| | | | - Pawel Kordowitzki
- Department for Basic and Preclinical Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Szosa Bydgoska 13, 87-100 Torun, Poland
| |
Collapse
|
12
|
Sasano T, Ihara K, Tanaka T, Furukawa T. Risk stratification of atrial fibrillation and stroke using single nucleotide polymorphism and circulating biomarkers. PLoS One 2023; 18:e0292118. [PMID: 37824462 PMCID: PMC10569505 DOI: 10.1371/journal.pone.0292118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/13/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common sustained arrhythmia, and it causes a high rate of complications such as stroke. It is known that AF begins as paroxysmal form and gradually progresses to persistent form, and sometimes it is difficult to identify paroxysmal AF (PAF) before having stroke. The aim of this study is to evaluate the risk of PAF and stroke using genetic analysis and circulating biomarkers. MATERIALS AND METHODS A total of 600 adult subjects were enrolled (300 from PAF and control groups). Peripheral blood was drawn to identify the genetic variation and biomarkers. Ten single nucleotide polymorphisms (SNPs) were analyzed, and circulating cell-free DNA (cfDNA) was measured from plasma. Four microRNAs (miR-99a-5p, miR-192-5p, miR-214-3p, and miR-342-5p) were quantified in serum using quantitative RT-PCR. RESULTS Genotyping identified 4 single nucleotide polymorphisms (SNPs) that were significantly associated with AF (rs6817105, rs3807989, rs10824026, and rs2106261), and the genetic risk score using 4 SNPs showed the area under the curve (AUC) of 0.631. Circulating miRNAs and cfDNA did not show significant differences between PAF and control groups. The concentration of cfDNA was significantly higher in patients with a history of stroke, and the AUC was 0.950 to estimate the association with stroke. CONCLUSION The risk of AF could be assessed by genetic risk score. Furthermore, the risk of stroke might be evaluated by plasma cfDNA level.
Collapse
Affiliation(s)
- Tetsuo Sasano
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kensuke Ihara
- Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Toshihiro Tanaka
- Bioresourse Research Center, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Department of Human Genetics and Disease Diversity, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tetsushi Furukawa
- Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
13
|
Jameson HS, Hanley A, Hill MC, Xiao L, Ye J, Bapat A, Ronzier E, Hall AW, Hucker WJ, Clauss S, Barazza M, Silber E, Mina J, Tucker NR, Mills RW, Dong JT, Milan DJ, Ellinor PT. Loss of the Atrial Fibrillation-Related Gene, Zfhx3, Results in Atrial Dilation and Arrhythmias. Circ Res 2023; 133:313-329. [PMID: 37449401 PMCID: PMC10527554 DOI: 10.1161/circresaha.123.323029] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/28/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND ZFHX3 (zinc finger homeobox 3), a gene that encodes a large transcription factor, is at the second-most significantly associated locus with atrial fibrillation (AF), but its function in the heart is unknown. This study aims to identify causative genetic variation related to AF at the ZFHX3 locus and examine the impact of Zfhx3 loss on cardiac function in mice. METHODS CRISPR-Cas9 genome editing, chromatin immunoprecipitation, and luciferase assays in pluripotent stem cell-derived cardiomyocytes were used to identify causative genetic variation related to AF at the ZFHX3 locus. Cardiac function was assessed by echocardiography, magnetic resonance imaging, electrophysiology studies, calcium imaging, and RNA sequencing in mice with heterozygous and homozygous cardiomyocyte-restricted Zfhx3 loss (Zfhx3 Het and knockout, respectively). Human cardiac single-nucleus ATAC (assay for transposase-accessible chromatin)-sequencing data was analyzed to determine which genes in atrial cardiomyocytes are directly regulated by ZFHX3. RESULTS We found single-nucleotide polymorphism (SNP) rs12931021 modulates an enhancer regulating ZFHX3 expression, and the AF risk allele is associated with decreased ZFHX3 transcription. We observed a gene-dose response in AF susceptibility with Zfhx3 knockout mice having higher incidence, frequency, and burden of AF than Zfhx3 Het and wild-type mice, with alterations in conduction velocity, atrial action potential duration, calcium handling and the development of atrial enlargement and thrombus, and dilated cardiomyopathy. Zfhx3 loss results in atrial-specific differential effects on genes and signaling pathways involved in cardiac pathophysiology and AF. CONCLUSIONS Our findings implicate ZFHX3 as the causative gene at the 16q22 locus for AF, and cardiac abnormalities caused by loss of cardiac Zfhx3 are due to atrial-specific dysregulation of pathways involved in AF susceptibility. Together, these data reveal a novel and important role for Zfhx3 in the control of cardiac genes and signaling pathways essential for normal atrial function.
Collapse
Affiliation(s)
- Heather S. Jameson
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Alan Hanley
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Matthew C. Hill
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Ling Xiao
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Jiangchuan Ye
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Aneesh Bapat
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Elsa Ronzier
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Amelia Weber Hall
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA
| | - William J. Hucker
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Sebastian Clauss
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine I, University Hospital Munich, Campus Grosshadern, Ludwig-Maximilians University Munich (LMU), 81377 Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site: Munich Heart Alliance, Munich, Germany
- Walter Brendel Centre of Experimental Medicine, University Hospital, LMU Munich, Germany
| | - Miranda Barazza
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Elizabeth Silber
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Julie Mina
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | | | - Robert W. Mills
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | | | | | - Patrick T. Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA
| |
Collapse
|
14
|
Wass SY, Smith JD, Chung MK. Connecting the Dots From GWAS to Function in Atrial Fibrillation for ZFHX3. Circ Res 2023; 133:330-332. [PMID: 37535755 PMCID: PMC10403289 DOI: 10.1161/circresaha.123.323281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Affiliation(s)
- Sojin Y Wass
- The Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute, and the Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, OH
| | - Jonathan D Smith
- The Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute, and the Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, OH
| | - Mina K Chung
- The Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute, and the Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, OH
| |
Collapse
|
15
|
Igarashi W, Takagi D, Okada D, Kobayashi D, Oka M, Io T, Ishii K, Ono K, Yamamoto H, Okamoto Y. Bioinformatic Identification of Potential RNA Alterations on the Atrial Fibrillation Remodeling from Human Pulmonary Veins. Int J Mol Sci 2023; 24:10501. [PMID: 37445678 DOI: 10.3390/ijms241310501] [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: 03/18/2023] [Revised: 05/16/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Atrial fibrillation (AF) is the most frequent persistent arrhythmia. Many genes have been reported as a genetic background for AF. However, most transcriptome analyses of AF are limited to the atrial samples and have not been evaluated by multiple cardiac regions. In this study, we analyzed the expression levels of protein-coding and long noncoding RNAs (lncRNAs) in six cardiac regions by RNA-seq. Samples were donated from six subjects with or without persistent AF for left atria, left atrial appendages, right atria, sinoatrial nodes, left ventricles, right ventricles, and pulmonary veins (PVs), and additional four right atrial appendages samples were collected from patients undergoing mitral valve replacement. In total, 23 AF samples were compared to 23 non-AF samples. Surprisingly, the most influenced heart region in gene expression by AF was the PV, not the atria. The ion channel-related gene set was significantly enriched upon analysis of these significant genes. In addition, some significant genes are cancer-related lncRNAs in PV in AF. A co-expression network analysis could detect the functional gene clusters. In particular, the cancer-related lncRNA, such as SAMMSON and FOXCUT, belong to the gene network with the cancer-related transcription factor FOXC1. Thus, they may also play an aggravating role in the pathogenesis of AF, similar to carcinogenesis. In the least, this study suggests that (1) RNA alteration is most intense in PVs and (2) post-transcriptional gene regulation by lncRNA may contribute to the progression of AF. Through the screening analysis across the six cardiac regions, the possibility that the PV region can play a role other than paroxysmal triggering in the pathogenesis of AF was demonstrated for the first time. Future research with an increase in the number of PV samples will lead to a novel understanding of the pathophysiology of AF.
Collapse
Affiliation(s)
- Wataru Igarashi
- Department of Cardiovascular Surgery, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | - Daichi Takagi
- Department of Cardiovascular Surgery, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | - Daigo Okada
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Shogoinkawahara-cho, Kyoto 606-8507, Japan
| | - Daiki Kobayashi
- Department of Cell Physiology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | - Miho Oka
- Research Department, Ono Pharmaceutical Co., Ltd., Kyutaromachi, Osaka 541-0056, Japan
| | - Toshiro Io
- Research Department, Ono Pharmaceutical Co., Ltd., Kyutaromachi, Osaka 541-0056, Japan
| | - Kuniaki Ishii
- Department of Pharmacology, Faculty of Medicine, Yamagata University, Iida-Nishi, Yamagata 990-9585, Japan
| | - Kyoichi Ono
- Department of Cell Physiology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | - Hiroshi Yamamoto
- Department of Cardiovascular Surgery, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | - Yosuke Okamoto
- Department of Cell Physiology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| |
Collapse
|
16
|
Patel KK, Venkatesan C, Abdelhalim H, Zeeshan S, Arima Y, Linna-Kuosmanen S, Ahmed Z. Genomic approaches to identify and investigate genes associated with atrial fibrillation and heart failure susceptibility. Hum Genomics 2023; 17:47. [PMID: 37270590 DOI: 10.1186/s40246-023-00498-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/31/2023] [Indexed: 06/05/2023] Open
Abstract
Atrial fibrillation (AF) and heart failure (HF) contribute to about 45% of all cardiovascular disease (CVD) deaths in the USA and around the globe. Due to the complex nature, progression, inherent genetic makeup, and heterogeneity of CVDs, personalized treatments are believed to be critical. To improve the deciphering of CVD mechanisms, we need to deeply investigate well-known and identify novel genes that are responsible for CVD development. With the advancements in sequencing technologies, genomic data have been generated at an unprecedented pace to foster translational research. Correct application of bioinformatics using genomic data holds the potential to reveal the genetic underpinnings of various health conditions. It can help in the identification of causal variants for AF, HF, and other CVDs by moving beyond the one-gene one-disease model through the integration of common and rare variant association, the expressed genome, and characterization of comorbidities and phenotypic traits derived from the clinical information. In this study, we examined and discussed variable genomic approaches investigating genes associated with AF, HF, and other CVDs. We collected, reviewed, and compared high-quality scientific literature published between 2009 and 2022 and accessible through PubMed/NCBI. While selecting relevant literature, we mainly focused on identifying genomic approaches involving the integration of genomic data; analysis of common and rare genetic variants; metadata and phenotypic details; and multi-ethnic studies including individuals from ethnic minorities, and European, Asian, and American ancestries. We found 190 genes associated with AF and 26 genes linked to HF. Seven genes had implications in both AF and HF, which are SYNPO2L, TTN, MTSS1, SCN5A, PITX2, KLHL3, and AGAP5. We listed our conclusion, which include detailed information about genes and SNPs associated with AF and HF.
Collapse
Affiliation(s)
- Kush Ketan Patel
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Cynthia Venkatesan
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Habiba Abdelhalim
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Saman Zeeshan
- Rutgers Cancer Institute of New Jersey, Rutgers University, 195 Little Albany St, New Brunswick, NJ, USA
| | - Yuichiro Arima
- Developmental Cardiology Laboratory, International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Kumamoto City, Kumamoto, Japan
| | - Suvi Linna-Kuosmanen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Zeeshan Ahmed
- Department of Genetics and Genome Sciences, UConn Health, 400 Farmington Ave, Farmington, CT, USA.
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers Biomedical and Health Sciences, 125 Paterson St, New Brunswick, NJ, USA.
| |
Collapse
|
17
|
Nakano Y. Genome and atrial fibrillation. J Arrhythm 2023; 39:303-309. [PMID: 37324776 PMCID: PMC10264727 DOI: 10.1002/joa3.12847] [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: 12/24/2022] [Revised: 03/11/2023] [Accepted: 03/27/2023] [Indexed: 06/17/2023] Open
Abstract
Atrial fibrillation (AF), the most common type of arrhythmia, can cause several adverse effects, such as stroke, heart failure, and cognitive dysfunction, also in addition to reducing quality of life and increasing mortality. Evidence suggests that AF is caused by a combination of genetic and clinical predispositions. In line with this, genetic studies on AF have progressed significantly through linkage studies, genome-wide association studies, use of polygenic risk scores, and studies on rare coding variations, gradually elucidating the relationship between genes and the pathogenesis and prognosis of AF. This article will review current trends in genetic analysis concerning AF.
Collapse
Affiliation(s)
- Yukiko Nakano
- Department of Cardiovascular MedicineHiroshima University Graduate School of Biomedical and Health SciencesHiroshimaJapan
| |
Collapse
|
18
|
Del Rocío Pérez Baca M, Jacobs EZ, Vantomme L, Leblanc P, Bogaert E, Dheedene A, De Cock L, Haghshenas S, Foroutan A, Levy MA, Kerkhof J, McConkey H, Chen CA, Batzir NA, Wang X, Palomares M, Carels M, Demaut B, Sadikovic B, Menten B, Yuan B, Vergult S, Callewaert B. A novel neurodevelopmental syndrome caused by loss-of-function of the Zinc Finger Homeobox 3 (ZFHX3) gene. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.22.23289895. [PMID: 37292950 PMCID: PMC10246128 DOI: 10.1101/2023.05.22.23289895] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Neurodevelopmental disorders (NDDs) result from impaired development and functioning of the brain. Here, we identify loss-of-function variation in ZFHX3 as a novel cause for syndromic intellectual disability (ID). ZFHX3, previously known as ATBF1, is a zinc-finger homeodomain transcription factor involved in multiple biological processes including cell differentiation and tumorigenesis. Through international collaboration, we collected clinical and morphometric data (Face2Gene) of 41 individuals with protein truncating variants (PTVs) or (partial) deletions of ZFHX3 . We used data mining, RNA and protein analysis to identify the subcellular localization and spatiotemporal expression of ZFHX3 in multiple in vitro models. We identified the DNA targets of ZFHX3 using ChIP seq. Immunoprecipitation followed by mass spectrometry indicated potential binding partners of endogenous ZFHX3 in neural stem cells that were subsequently confirmed by reversed co-immunoprecipitation and western blot. We evaluated a DNA methylation profile associated with ZFHX3 haploinsufficiency using DNA methylation analysis on whole blood extracted DNA of six individuals with ZFHX3 PTVs and four with a (partial) deletion of ZFHX3 . A reversed genetic approach characterized the ZFHX3 orthologue in Drosophila melanogaster . Loss-of-function variation of ZFHX3 consistently associates with (mild) ID and/or behavioural problems, postnatal growth retardation, feeding difficulties, and recognizable facial characteristics, including the rare occurrence of cleft palate. Nuclear abundance of ZFHX3 increases during human brain development and neuronal differentiation in neural stem cells and SH-SY5Y cells, ZFHX3 interacts with the chromatin remodelling BRG1/Brm-associated factor complex and the cleavage and polyadenylation complex. In line with a role for chromatin remodelling, ZFHX3 haploinsufficiency associates with a specific DNA methylation profile in leukocyte-derived DNA. The target genes of ZFHX3 are implicated in neuron and axon development. In Drosophila melanogaster , z fh2, considered to be the ZFHX3 orthologue, is expressed in the third instar larval brain. Ubiquitous and neuron-specific knockdown of zfh2 results in adult lethality underscoring a key role for zfh2 in development and neurodevelopment. Interestingly, ectopic expression of zfh2 as well as ZFHX3 in the developing wing disc results in a thoracic cleft phenotype. Collectively, our data shows that loss-of-function variants in ZFHX3 are a cause of syndromic ID, that associates with a specific DNA methylation profile. Furthermore, we show that ZFHX3 participates in chromatin remodelling and mRNA processing.
Collapse
|
19
|
Huang PS, Cheng JF, Li GW, Chuang EY, Chen JJ, Chiu FC, Wu CK, Wang YC, Hwang JJ, Tsai CT. Copy number variation of gasdermin D gene is associated with atrial fibrillation-related thromboembolic stroke. Europace 2023; 25:euad103. [PMID: 37083255 PMCID: PMC10228625 DOI: 10.1093/europace/euad103] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 03/30/2023] [Indexed: 04/22/2023] Open
Abstract
AIMS Atrial fibrillation (AF) is one of the major causes of ischaemic stroke. In addition to clinical risk evaluated by the CHA2DS2-VASC score, the impact of genetic factors on the risk of AF-related thromboembolic stroke has been largely unknown. We found several copy number variations (CNVs) in novel genes that were associated with thromboembolic stroke risk in our AF patients by genome-wide approach. Among them, the gasdermin D (GSDMD) gene was related to inflammation. We aimed to test whether GSDMD deletion was associated with AF-related stroke. METHODS AND RESULTS A total of 400 patients with documented non-familial AF were selected, of which 100 patients were diagnosed with ischaemic stroke. The baseline characteristics of age, sex, valvular heart disease, coronary artery disease, heart failure, and CHA2DS2-VASc scores were not statistically different between cases and controls. We found that individuals who carried GSDMD homozygous deletion genotype had a higher risk for ischaemic stroke (odds ratio 2.195; 95% confidence interval, 1.24-3.90; P = 0.007), even adjusted by CHA2DS2-VASc scores. We also validated the association of GSDMD with AF stroke in a large Caucasian population (UK Biobank). CONCLUSION We found a link between the homozygous deletion of the GSDMD gene and an increased risk of stroke in patients with AF. This may implicate the use of therapy targeting GSDMD in the prevention of ischaemic stroke for AF patients.
Collapse
Affiliation(s)
- Pang-Shuo Huang
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, Yun-Lin, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Taiwan University HospitalNo. 7, Chung-Shan South Road, Taipei 100, Taiwan
- Cardiovascular Center, National Taiwan University Hospital, No.7, Zhongshan S. Rd., Zhongzheng District, Taipei City 10002, Taiwan
| | - Jen-Fang Cheng
- Division of Cardiology, Department of Internal Medicine, National Taiwan University HospitalNo. 7, Chung-Shan South Road, Taipei 100, Taiwan
- Cardiovascular Center, National Taiwan University Hospital, No.7, Zhongshan S. Rd., Zhongzheng District, Taipei City 10002, Taiwan
- Division of Multidisciplinary medicine, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Guan-Wei Li
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Eric Y Chuang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
- Bioinformatics and Biostatistics Core, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
- Research and Development Center for Medical Devices, National Taiwan University, Taipei, Taiwan
| | - Jien-Jiun Chen
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, Yun-Lin, Taiwan
| | - Fu-Chun Chiu
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, Yun-Lin, Taiwan
| | - Cho-Kai Wu
- Division of Cardiology, Department of Internal Medicine, National Taiwan University HospitalNo. 7, Chung-Shan South Road, Taipei 100, Taiwan
- Cardiovascular Center, National Taiwan University Hospital, No.7, Zhongshan S. Rd., Zhongzheng District, Taipei City 10002, Taiwan
| | - Yi-Chih Wang
- Division of Cardiology, Department of Internal Medicine, National Taiwan University HospitalNo. 7, Chung-Shan South Road, Taipei 100, Taiwan
- Cardiovascular Center, National Taiwan University Hospital, No.7, Zhongshan S. Rd., Zhongzheng District, Taipei City 10002, Taiwan
| | - Juey-Jen Hwang
- Division of Cardiology, Department of Internal Medicine, National Taiwan University HospitalNo. 7, Chung-Shan South Road, Taipei 100, Taiwan
- Cardiovascular Center, National Taiwan University Hospital, No.7, Zhongshan S. Rd., Zhongzheng District, Taipei City 10002, Taiwan
| | - Chia-Ti Tsai
- Division of Cardiology, Department of Internal Medicine, National Taiwan University HospitalNo. 7, Chung-Shan South Road, Taipei 100, Taiwan
- Cardiovascular Center, National Taiwan University Hospital, No.7, Zhongshan S. Rd., Zhongzheng District, Taipei City 10002, Taiwan
| |
Collapse
|
20
|
Wang MF, Xue C, Shi SY, Yang L, Zhu ZY, Li JJ. Gene Polymorphism and Recurrent Atrial Fibrillation after Catheter Ablation: A Comprehensive Review. Rev Cardiovasc Med 2023; 24:119. [PMID: 39076272 PMCID: PMC11273024 DOI: 10.31083/j.rcm2404119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/11/2022] [Accepted: 12/19/2022] [Indexed: 08/15/2023] Open
Abstract
Atrial fibrillation (AF) is one of the most common cardiac arrhythmias, but its pathogenesis is still poorly understood. Catheter ablation is one of the most effective treatments for AF, but recurrence after ablation remains a challenge. There has been much research into the association of AF recurrence with several factors, including genetics. Over the past decade or so, significant advances have been made in the genetic architecture of atrial fibrillation. Genome-wide association studies (GWAS) have identified over 100 loci for genetic variants associated with atrial fibrillation. However, there is relatively little information on the systematic assessment of the genes related to AF recurrence after ablation. In this review article, we highlight the value of genetic polymorphisms in atrial fibrillation recurrence after catheter ablation and their potential mechanisms in the recurrence process to enhance our understanding of atrial fibrillation recurrence and contribute to individualized treatment strategies for patients with AF.
Collapse
Affiliation(s)
- Meng-Fei Wang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, 213000 Changzhou, Jiangsu, China
| | - Cong Xue
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, 213000 Changzhou, Jiangsu, China
| | - Shun-Yi Shi
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, 213000 Changzhou, Jiangsu, China
| | - Ling Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, 213000 Changzhou, Jiangsu, China
| | - Zhen-Yan Zhu
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, 213000 Changzhou, Jiangsu, China
| | - Jian-Jun Li
- State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
| |
Collapse
|
21
|
Casalino-Matsuda SM, Chen F, Gonzalez-Gonzalez FJ, Matsuda H, Nair A, Abdala-Valencia H, Budinger GRS, Dong JT, Beitel GJ, Sporn PHS. Myeloid Zfhx3 Deficiency Protects Against Hypercapnia-induced Suppression of Host Defense Against Influenza A Virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.28.530480. [PMID: 36909510 PMCID: PMC10002734 DOI: 10.1101/2023.02.28.530480] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Hypercapnia, elevation of the partial pressure of CO 2 in blood and tissues, is a risk factor for mortality in patients with severe acute and chronic lung diseases. We previously showed that hypercapnia inhibits multiple macrophage and neutrophil antimicrobial functions, and that elevated CO 2 increases the mortality of bacterial and viral pneumonia in mice. Here, we show that normoxic hypercapnia downregulates innate immune and antiviral gene programs in alveolar macrophages (AMØs). We also show that zinc finger homeobox 3 (Zfhx3), mammalian ortholog of zfh2, which mediates hypercapnic immune suppression in Drosophila , is expressed in mouse and human MØs. Deletion of Zfhx3 in the myeloid lineage blocked the suppressive effect of hypercapnia on immune gene expression in AMØs and decreased viral replication, inflammatory lung injury and mortality in hypercapnic mice infected with influenza A virus. Our results establish Zfhx3 as the first known mammalian mediator of CO 2 effects on immune gene expression and lay the basis for future studies to identify therapeutic targets to interrupt hypercapnic immunosuppression in patients with advanced lung diseases. Graphical abstract
Collapse
|
22
|
Roh J, Im M, Kang J, Youn B, Kim W. Long non-coding RNA in glioma: novel genetic players in temozolomide resistance. Anim Cells Syst (Seoul) 2023; 27:19-28. [PMID: 36819921 PMCID: PMC9937017 DOI: 10.1080/19768354.2023.2175497] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
Glioma is the most common primary malignant brain tumor in adults and accounts for approximately 80% of brain and central nervous system tumors. In 2021, the World Health Organization (WHO) published a new taxonomy for glioma based on its histological features and molecular alterations. Isocitrate dehydrogenase (IDH) catalyzes the decarboxylation of isocitrate, a critical metabolic reaction in energy generation in cells. Mutations in the IDH genes interrupt cell differentiation and serve as molecular biomarkers that can be used to classify gliomas. For example, the mutant IDH is widely detected in low-grade gliomas, whereas the wild type is in high-grade ones, including glioblastomas. Long non-coding RNAs (lncRNAs) are epigenetically involved in gene expression and contribute to glioma development. To investigate the potential use of lncRNAs as biomarkers, we examined lncRNA dysregulation dependent on the IDH mutation status. We found that several lncRNAs, namely, AL606760.2, H19, MALAT1, PVT1 and SBF2-AS1 may function as glioma risk factors, whereas AC068643.1, AC079228.1, DGCR5, FAM13A-AS1, HAR1A and WDFY3-AS2 may have protective effects. Notably, H19, MALAT1, PVT1, and SBF2-AS1 have been associated with temozolomide resistance in glioma patients. This review study suggests that targeting glioma-associated lncRNAs might aid the treatment of glioma.
Collapse
Affiliation(s)
- Jungwook Roh
- Department of Science Education, Korea National University of Education, Cheongju-si, Republic of Korea
| | - Mijung Im
- Department of Science Education, Korea National University of Education, Cheongju-si, Republic of Korea
| | - JiHoon Kang
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory, Emory University School of Medicine, Atlanta, GA, USA
| | - BuHyun Youn
- Department of Biological Sciences, Pusan National University, Busan, Republic of Korea, BuHyun Youn Department of Biological Sciences, Pusan National University, Busandaehak-ro 63beon-gil 2, Geumjeong-gu, Busan46241, Republic of Korea; Wanyeon Kim Department of Biology Education, Korea National University of Education, 250 Taeseongtabyeon-ro, Gangnae-myeon, Heungdeok-gu, Cheongju-si, Chungbuk28173, Republic of Korea
| | - Wanyeon Kim
- Department of Science Education, Korea National University of Education, Cheongju-si, Republic of Korea,Department of Biology Education, Korea National University of Education, Cheongju-si, Republic of Korea, BuHyun Youn Department of Biological Sciences, Pusan National University, Busandaehak-ro 63beon-gil 2, Geumjeong-gu, Busan46241, Republic of Korea; Wanyeon Kim Department of Biology Education, Korea National University of Education, 250 Taeseongtabyeon-ro, Gangnae-myeon, Heungdeok-gu, Cheongju-si, Chungbuk28173, Republic of Korea
| |
Collapse
|
23
|
Kao YH, Chen YJ, Higa S, Chattipakorn N, Santulli G. Editorial: Transcription factors and arrhythmogenesis. Front Physiol 2023; 14:1169747. [PMID: 36926195 PMCID: PMC10011700 DOI: 10.3389/fphys.2023.1169747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 03/08/2023] Open
Affiliation(s)
- Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Internal Medicine, Division of Cardiovascular Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Satoshi Higa
- Cardiac Electrophysiology and Pacing Laboratory, Division of Cardiovascular Medicine, Makiminato Central Hospital, Urasoe, Japan
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Gaetano Santulli
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY, United States.,Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Department of Molecular Pharmacology, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
24
|
Crespo-García T, Cámara-Checa A, Dago M, Rubio-Alarcón M, Rapún J, Tamargo J, Delpón E, Caballero R. Regulation of cardiac ion channels by transcription factors: Looking for new opportunities of druggable targets for the treatment of arrhythmias. Biochem Pharmacol 2022; 204:115206. [PMID: 35963339 DOI: 10.1016/j.bcp.2022.115206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/04/2022] [Accepted: 08/05/2022] [Indexed: 11/29/2022]
Abstract
Cardiac electrical activity is governed by different ion channels that generate action potentials. Acquired or inherited abnormalities in the expression and/or function of ion channels usually result in electrophysiological changes that can cause cardiac arrhythmias. Transcription factors (TFs) control gene transcription by binding to specific DNA sequences adjacent to target genes. Linkage analysis, candidate-gene screening within families, and genome-wide association studies have linked rare and common genetic variants in the genes encoding TFs with genetically-determined cardiac arrhythmias. Besides its critical role in cardiac development, recent data demonstrated that they control cardiac electrical activity through the direct regulation of the expression and function of cardiac ion channels in adult hearts. This narrative review summarizes some studies showing functional data on regulation of the main human atrial and ventricular Na+, Ca2+, and K+ channels by cardiac TFs such as Pitx2c, Tbx20, Tbx5, Zfhx3, among others. The results have improved our understanding of the mechanisms regulating cardiac electrical activity and may open new avenues for therapeutic interventions in cardiac acquired or inherited arrhythmias through the identification of TFs as potential drug targets. Even though TFs have for a long time been considered as 'undruggable' targets, advances in structural biology have led to the identification of unique pockets in TFs amenable to be targeted with small-molecule drugs or peptides that are emerging as novel therapeutic drugs.
Collapse
Affiliation(s)
- T Crespo-García
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain
| | - A Cámara-Checa
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain
| | - M Dago
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain
| | - M Rubio-Alarcón
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain
| | - J Rapún
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain
| | - J Tamargo
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain
| | - E Delpón
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain.
| | - R Caballero
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain
| | -
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain
| |
Collapse
|
25
|
Li Q, Lei S, Luo X, He J, Fang Y, Yang H, Liu Y, Deng CY, Wu S, Xue YM, Rao F. Construction of Prediction Model for Atrial Fibrillation with Valvular Heart Disease Based on Machine Learning. Rev Cardiovasc Med 2022; 23:247. [PMID: 39076905 PMCID: PMC11266776 DOI: 10.31083/j.rcm2307247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/31/2022] [Accepted: 06/10/2022] [Indexed: 07/31/2024] Open
Abstract
Background Valvular heart disease (VHD) is a major precipitating factor of atrial fibrillation (AF) that contributes to decreased cardiac function, heart failure, and stroke. Stroke induced by VHD combined with atrial fibrillation (AF-VHD) is a much more serious condition in comparison to VHD alone. The aim of this study was to explore the molecular mechanism governing VHD progression and to provide candidate treatment targets for AF-VHD. Methods Four public mRNA microarray datasets were downloaded and differentially expressed genes (DEGs) screening was performed. Weighted gene correlation network analysis was carried out to detect key modules and explore their relationships and disease status. Candidate hub signature genes were then screened within the key module using machine learning methods. The receiver operating characteristic curve and nomogram model analysis were used to determine the potential clinical significance of the hub genes. Subsequently, target gene protein levels in independent human atrial tissue samples were detected using western blotting. Specific expression analysis of the hub genes in the tissue and cell samples was performed using single-cell sequencing analysis in the Human Protein Atlas tool. Results A total of 819 common DEGs in combined datasets were screened. Fourteen modules were identified using the cut tree dynamic function. The cyan and purple modules were considered the most clinically significant for AF-VHD. Then, 25 hub genes in the cyan and purple modules were selected for further analysis. The pathways related to dilated cardiomyopathy, hypertrophic cardiomyopathy, and heart contraction were concentrated in the purple and cyan modules of the AF-VHD. Genes of importance (CSRP3, MCOLN3, SLC25A5, and FIBP) were then identified based on machine learning. Of these, CSRP3 had a potential clinical significance and was specifically expressed in the heart tissue. Conclusions The identified genes may play critical roles in the pathophysiological process of AF-VHD, providing new insights into VHD development to AF and helping to determine potential biomarkers and therapeutic targets for treating AF-VHD.
Collapse
Affiliation(s)
- Qiaoqiao Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
- Research Center of Medical Sciences, Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
| | - Shenghong Lei
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
- Research Center of Medical Sciences, Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
| | - Xueshan Luo
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
- Research Center of Medical Sciences, Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
| | - Jintao He
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
- Research Center of Medical Sciences, Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
| | - Yuan Fang
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
- Research Center of Medical Sciences, Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
| | - Hui Yang
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
- Research Center of Medical Sciences, Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
| | - Yang Liu
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
- Research Center of Medical Sciences, Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
| | - Chun-Yu Deng
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
- Research Center of Medical Sciences, Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
| | - Shulin Wu
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
- Research Center of Medical Sciences, Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
| | - Yu-Mei Xue
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
- Research Center of Medical Sciences, Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
| | - Fang Rao
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
- Research Center of Medical Sciences, Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, Guangdong, China
| |
Collapse
|
26
|
Pirruccello JP, Di Achille P, Nauffal V, Nekoui M, Friedman SF, Klarqvist MDR, Chaffin MD, Weng LC, Cunningham JW, Khurshid S, Roselli C, Lin H, Koyama S, Ito K, Kamatani Y, Komuro I, Jurgens SJ, Benjamin EJ, Batra P, Natarajan P, Ng K, Hoffmann U, Lubitz SA, Ho JE, Lindsay ME, Philippakis AA, Ellinor PT. Genetic analysis of right heart structure and function in 40,000 people. Nat Genet 2022; 54:792-803. [PMID: 35697867 PMCID: PMC10313645 DOI: 10.1038/s41588-022-01090-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/26/2022] [Indexed: 01/29/2023]
Abstract
Congenital heart diseases often involve maldevelopment of the evolutionarily recent right heart chamber. To gain insight into right heart structure and function, we fine-tuned deep learning models to recognize the right atrium, right ventricle and pulmonary artery, measuring right heart structures in 40,000 individuals from the UK Biobank with magnetic resonance imaging. Genome-wide association studies identified 130 distinct loci associated with at least one right heart measurement, of which 72 were not associated with left heart structures. Loci were found near genes previously linked with congenital heart disease, including NKX2-5, TBX5/TBX3, WNT9B and GATA4. A genome-wide polygenic predictor of right ventricular ejection fraction was associated with incident dilated cardiomyopathy (hazard ratio, 1.33 per standard deviation; P = 7.1 × 10-13) and remained significant after accounting for a left ventricular polygenic score. Harnessing deep learning to perform large-scale cardiac phenotyping, our results yield insights into the genetic determinants of right heart structure and function.
Collapse
Affiliation(s)
- James P Pirruccello
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Paolo Di Achille
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Victor Nauffal
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Mahan Nekoui
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Samuel F Friedman
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marcus D R Klarqvist
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mark D Chaffin
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lu-Chen Weng
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonathan W Cunningham
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Shaan Khurshid
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Carolina Roselli
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Honghuang Lin
- Framingham Heart Study, Boston University and National Heart, Lung, and Blood Institute, Framingham, MA, USA
- Division of Clinical Informatics, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Satoshi Koyama
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Laboratory for Cardiovascular Genomics and Informatics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Kaoru Ito
- Laboratory for Cardiovascular Genomics and Informatics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Yoichiro Kamatani
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sean J Jurgens
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Emelia J Benjamin
- Framingham Heart Study, Boston University and National Heart, Lung, and Blood Institute, Framingham, MA, USA
- Department of Medicine, Cardiology and Preventive Medicine Sections, Boston University School of Medicine, Boston, MA, USA
- Epidemiology Department, Boston University School of Public Health, Boston, MA, USA
| | - Puneet Batra
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Pradeep Natarajan
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Udo Hoffmann
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Steven A Lubitz
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Jennifer E Ho
- Harvard Medical School, Boston, MA, USA
- CardioVascular Institute and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Mark E Lindsay
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Thoracic Aortic Center, Massachusetts General Hospital, Boston, MA, USA
| | | | - Patrick T Ellinor
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
27
|
Gao J, Bai Y, Ji H. Genetically Predicted Atrial Fibrillation and Valvular Heart Disease: A Two-Sample Mendelian Randomization Study. Front Cardiovasc Med 2022; 9:845734. [PMID: 35419428 PMCID: PMC8996053 DOI: 10.3389/fcvm.2022.845734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/18/2022] [Indexed: 11/22/2022] Open
Abstract
Background Previous studies have found atrial fibrillation (AF) is associated with valvular heart disease (VHD). However, whether there is a causal relationship between these two diseases or it is just a result of bias caused by confounding factors is uncertain. This study aims to examine the potential causal association between AF and VHD by using Mendelian randomization. Methods In order to examine the causal relationship between AF and VHD, we performed a two-sample Mendelian randomization study by collecting exposure and outcome data from genome-wide association study (GWAS) datasets. We utilized data from FinnGen project (FinnGen, 11,258 cases for VHD including rheumatic fever, 3,108 cases for non-rheumatic VHD, and 75,137 cases for participants) and European Bio-informatics Institute database (EBI, 55,114 cases for AF and 482,295 cases for participants). Inverse-variance weighted (IVW), MR-Egger, and weighted median approaches were performed to estimate the causal effect. Results The Mendelian randomization analysis indicated that AF increased the risk of VHD by all three MR methods [For VHD including rheumatic fever: IVW, odds ratio (OR) = 1.255; 95% confidence interval (CI), 1.191~1.322; p = 1.23 × 10−17; Weighted median, OR = 1.305, 95% CI, 1.216~1.400, p = 1.57 × 10−13; MR-Egger, OR = 1.250, 95% CI, 1.137~1.375, p = 1.69 × 10−5; For non-rheumatic VHD: IVW, OR = 1.267; 95% CI, 1.169~1.372; p = 6.73 × 10−9; Weighted median, OR = 1.400; 95% CI, 1.232~1.591; p = 2.40 × 10−7; MR-Egger, OR = 1.308; 95% CI, 1.131~1.513; p = 5.34 × 10−4]. After the one outlier SNP was removed by heterogeneity test, the results remained the same. No horizontal pleiotropic effects were observed between AF and VHD. Conclusions Our study provides strong evidence of a causal relationship between AF and VHD. Early intervention for AF patients may reduce the risk of developing into VHD.
Collapse
Affiliation(s)
- Jie Gao
- Department of Anesthesiology, Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yulin Bai
- Department of Cardiac Surgery, The 7th People's Hospital of Zhengzhou, Zhengzhou, China
| | - Hongwen Ji
- Department of Anesthesiology, Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- *Correspondence: Hongwen Ji
| |
Collapse
|
28
|
Zigova M, Petrejèíková E, Blašèáková M, Kmec J, Bernasovská J, Boroòová I, Kmec M. Genetic targets in the management of atrial fibrillation in patients with cardiomyopathy. JOURNAL OF THE PRACTICE OF CARDIOVASCULAR SCIENCES 2022. [DOI: 10.4103/jpcs.jpcs_65_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
29
|
Association between ZFHX3 and PRRX1 Polymorphisms and Atrial Fibrillation Susceptibility from Meta-Analysis. Int J Hypertens 2021; 2021:9423576. [PMID: 34950514 PMCID: PMC8692054 DOI: 10.1155/2021/9423576] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 10/15/2021] [Indexed: 11/17/2022] Open
Abstract
Background Atrial fibrillation (AF) is a common, sustained cardiac arrhythmia. Recent studies have reported an association between ZFHX3/PRRX1 polymorphisms and AF. In this study, a meta-analysis was conducted to confirm these associations. Objective and Methods. The PubMed, Embase, and Wanfang databases were searched, covering all publications before July 20, 2020. Results Overall, seven articles including 3,674 cases and 8,990 healthy controls for ZFHX3 rs2106261 and 1045 cases and 1407 controls for PRRX1 rs3903239 were included. The odds ratio (OR) (95% confidence interval (CI)) was used to assess the associations. Publication bias was calculated using Egger's and Begg's tests. We found that the ZFHX3 rs2106261 polymorphism increased AF risk in Asians (for example, allelic contrast: OR [95% CI]: 1.39 [1.31–1.47], P < 0.001). Similarly, strong associations were detected through stratified analysis using source of control and genotype methods (for example, allelic contrast: OR [95% CI]: 1.51 [1.38–1.64], P < 0.001 for HB; OR [95% CI]: 1.31 [1.21–1.41], P < 0.001 for PB; OR [95% CI]: 1.55 [1.33–1.80], P < 0.001 for TaqMan; and OR [95% CI]: 1.31 [1.21–1.41], P < 0.001 for high-resolution melt). In contrast, an inverse relationship was observed between the PRRX1 rs3903239 polymorphism and AF risk (C-allele vs. T-allele: OR [95% CI]: 0.83 [0.77–0.99], P=0.036; CT vs. TT: OR [95% CI]: 0.79 [0.67–0.94], P=0.006). No obvious evidence of publication bias was observed. Conclusions In summary, our study suggests that the ZFHX3 rs2106261 and PRRX1 rs3903239 polymorphisms are associated with AF risk, and larger case-controls must be carried out to confirm the abovementioned conclusions.
Collapse
|
30
|
Wei Y, Wang L, Lin C, Xie Y, Bao Y, Luo Q, Zhang N. Association between the rs2106261 polymorphism in the zinc finger homeobox 3 gene and risk of atrial fibrillation: Evidence from a PRISMA-compliant meta-analysis. Medicine (Baltimore) 2021; 100:e27749. [PMID: 34889223 PMCID: PMC8663867 DOI: 10.1097/md.0000000000027749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 10/26/2021] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION Previous genome-wide studies have identified an association between the rs2106261 single-nucleotide polymorphism (SNP) in the zinc finger homeobox 3 (ZFHX3) gene and an increased risk of atrial fibrillation (AF). However, this association remains controversial, since conflicting results have been reported in previous studies. We aimed to investigate the association between the ZFHX3 rs2106261 polymorphism and susceptibility to AF. METHODS A comprehensive literature search, of articles written in either English or Chinese, was conducted on various databases, including PubMed, Embase, Web of Science, the Cochrane library, Wan Fang, and CNKI, for studies performed up to August 1, 2020. Data were abstracted and pooled using Stata 14.0 software. A meta-analysis was performed on all selected studies based on ZFHX3 rs2106261 polymorphism genotypes. RESULTS Nine studies, including 10,107 cases and 58,663 controls, were analyzed in the meta-analysis. In the overall population, a significant association was found between AF and the T-allelic ZFHX 3 rs2106261 SNP (odds ratio [OR] = 1.32, 95% confidence interval [CI] 1.19-1.46). In subgroup analysis, a significant association between the T-allele of rs7193343 and risk of AF in Caucasian (OR = 1.23, 95% CI 1.10-1.37) and Asian subgroups (OR = 1.58, 95% CI 1.32-1.89) was observed. However, no statistically significant association was found in African populations (OR = 1.06, 95% CI 0.95-1.19). CONCLUSION The genetic variant rs2106261 SNP is associated with susceptibility to AF in Caucasian and Asian individuals, with Asian samples showing a stronger association. However, based on the current evidence, no association was found in African samples. Future studies, with larger sample sizes and multiple ethnicities, are still necessary.
Collapse
|
31
|
Rubio-Alarcón M, Cámara-Checa A, Dago M, Crespo-García T, Nieto-Marín P, Marín M, Merino JL, Toquero J, Salguero-Bodes R, Tamargo J, Cebrián J, Delpón E, Caballero R. Zfhx3 Transcription Factor Represses the Expression of SCN5A Gene and Decreases Sodium Current Density (I Na). Int J Mol Sci 2021; 22:13031. [PMID: 34884836 PMCID: PMC8657907 DOI: 10.3390/ijms222313031] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 02/02/2023] Open
Abstract
The ZFHX3 and SCN5A genes encode the zinc finger homeobox 3 (Zfhx3) transcription factor (TF) and the human cardiac Na+ channel (Nav1.5), respectively. The effects of Zfhx3 on the expression of the Nav1.5 channel, and in cardiac excitability, are currently unknown. Additionally, we identified three Zfhx3 variants in probands diagnosed with familial atrial fibrillation (p.M1260T) and Brugada Syndrome (p.V949I and p.Q2564R). Here, we analyzed the effects of native (WT) and mutated Zfhx3 on Na+ current (INa) recorded in HL-1 cardiomyocytes. ZFHX3 mRNA can be detected in human atrial and ventricular samples. In HL-1 cardiomyocytes, transfection of Zfhx3 strongly reduced peak INa density, while the silencing of endogenous expression augmented it (from -65.9 ± 8.9 to -104.6 ± 10.8 pA/pF; n ≥ 8, p < 0.05). Zfhx3 significantly reduced the transcriptional activity of human SCN5A, PITX2, TBX5, and NKX25 minimal promoters. Consequently, the mRNA and/or protein expression levels of Nav1.5 and Tbx5 were diminished (n ≥ 6, p < 0.05). Zfhx3 also increased the expression of Nedd4-2 ubiquitin-protein ligase, enhancing Nav1.5 proteasomal degradation. p.V949I, p.M1260T, and p.Q2564R Zfhx3 produced similar effects on INa density and time- and voltage-dependent properties in WT. WT Zfhx3 inhibits INa as a result of a direct repressor effect on the SCN5A promoter, the modulation of Tbx5 increasing on the INa, and the increased expression of Nedd4-2. We propose that this TF participates in the control of cardiac excitability in human adult cardiac tissue.
Collapse
Affiliation(s)
- Marcos Rubio-Alarcón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - Anabel Cámara-Checa
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - María Dago
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - Teresa Crespo-García
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - Paloma Nieto-Marín
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - María Marín
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - José Luis Merino
- Department of Cardiology, Hospital Universitario La Paz, Instituto de Investigación Sanitaria la Paz, CIBERCV, 28046 Madrid, Spain;
| | - Jorge Toquero
- Department of Cardiology, Hospital Universitario Puerta de Hierro, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, CIBERCV, Majadahonda, 28222 Madrid, Spain;
| | - Rafael Salguero-Bodes
- Department of Cardiology, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre, CIBERCV, 28041 Madrid, Spain;
| | - Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - Jorge Cebrián
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - Eva Delpón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - Ricardo Caballero
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| |
Collapse
|
32
|
Linz D, Verheule S, Isaacs A, Schotten U. Considerations for the Assessment of Substrates, Genetics and Risk Factors in Patients with Atrial Fibrillation. Arrhythm Electrophysiol Rev 2021; 10:132-139. [PMID: 34777816 PMCID: PMC8576487 DOI: 10.15420/aer.2020.51] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 06/16/2021] [Indexed: 12/25/2022] Open
Abstract
Successful translation of research focussing on atrial arrhythmogenic mechanisms has potential to provide a mechanism-tailored classification and to support personalised treatment approaches in patients with AF. The clinical uptake and clinical implementation of new diagnostic techniques and treatment strategies require translational research approaches on various levels. Diagnostic translation involves the development of clinical diagnostic tools. Additionally, multidisciplinary teams are required for collaborative translation to describe genetic mechanisms, molecular pathways, electrophysiological characteristics and concomitant risk factors. In this article, current approaches for AF substrate characterisation, analysis of genes potentially involved in AF and strategies for AF risk factor assessment are summarised. The authors discuss challenges and obstacles to clinical translation and implementation into clinical practice.
Collapse
Affiliation(s)
- Dominik Linz
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine and Life Sciences, Maastricht University and Maastricht University Medical Center+, Maastricht, the Netherlands.,Department of Cardiology, Radboud University Medical Centre, Nijmegen, the Netherlands.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.,Centre for Heart Rhythm Disorders, University of Adelaide and Royal Adelaide Hospital, Adelaide, Australia
| | - Sander Verheule
- Department of Physiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Aaron Isaacs
- Department of Physiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Ulrich Schotten
- Department of Physiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
33
|
Fu T, Chen M, Xu L, Gong J, Zheng J, Zhang F, Ji N. Association of the MYH6 Gene Polymorphism with the Risk of Atrial Fibrillation and Warfarin Anticoagulation Therapy. Genet Test Mol Biomarkers 2021; 25:590-599. [PMID: 34515533 DOI: 10.1089/gtmb.2021.0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Objective: To study the associations of single nucleotide polymorphisms (SNP) of the myosin heavy chain 6 (MYH6) gene with the risk of atrial fibrillation (AF) and warfarin anticoagulation therapy. Methods: Sanger sequencing was employed to analyze the genotypes of the MYH6 gene's rs28730771, rs365990, and rs2277473 loci in 243 AF patients and 243 non-AF patients (control group) selected according to the age and sex of AF patients at a 1:1 ratio. A multiple logistic regression analysis was used to analyze the risk factors in AF. SHEsis was adopted to analyze the association between rs28730771, rs365990, rs2277473 haplotypes and susceptibility to AF. The average weekly doses of warfarin administered to AF patients with different genotypes were compared. Results: The T allele at rs28730771 of the MYH6 gene (odds ratio [OR] = 2.82, 95% confidence interval [CI]: 1.73-4.59, p < 0.01), the G allele at rs365990 (OR = 1.65, 95% CI: 1.22-2.24, p < 0.01) and the T allele at rs2277473 (OR = 1.91, 95% CI: 1.25-2.91, p < 0.01) were significantly associated with an elevated risk of AF. The results of a logistic regression analysis demonstrated that hypertension, smoking, drinking, family history of stroke, as well as the genotypes at the rs28730771, rs365990, and rs2277473 loci were all risk factors in AF (p < 0.05). The CAG haplotype for the three SNPs was associated with a reduced risk of AF susceptibility (OR = 0.61, 95% CI: 0.46-0.81, p < 0.01), and the CGG haplotype was related to an increased risk of AF (OR = 1.49, 95% CI: 1.07-2.06, p = 0.02). The doses of warfarin used in AF patients with different genotypes at the MYH6 rs28730771, rs365990, and rs2277473 loci were significantly different (p < 0.05). Conclusion: The three SNPs (rs28730771, rs365990, and rs2277473) of the MYH6 gene loci were significantly associated with the risk of AF susceptibility and the dose of warfarin anticoagulant therapy.
Collapse
Affiliation(s)
- Ting Fu
- Department of Cardiology, Yiwu Central Hospital, Yiwu, China
| | - Mengyan Chen
- Department of Cardiology, Yiwu Central Hospital, Yiwu, China
| | - Lei Xu
- Department of Cardiology, Yiwu Central Hospital, Yiwu, China
| | - Jianping Gong
- Department of Cardiology, Yiwu Central Hospital, Yiwu, China
| | - Juanqing Zheng
- Department of Cardiology, Yiwu Central Hospital, Yiwu, China
| | - Fen Zhang
- Department of Cardiology, Jinhua People's Hospital, Jinhua, China
| | - Ningning Ji
- Department of Cardiology, Yiwu Central Hospital, Yiwu, China
| |
Collapse
|
34
|
Detection of Subclinical Paroxysmal Atrial Fibrillation and Its Correlation with Candidate Genes in Patients with Cryptogenic Ischemic Stroke and TIA. ACTA MEDICA MARTINIANA 2021. [DOI: 10.2478/acm-2021-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Introduction: Cardioembolic etiology is assumed to be the most frequent cause of cryptogenic strokes. The detection of subclinical paroxysmal atrial fibrillation (AF) is important in the correct choice of preventive treatment. The aim of this prospective study was to detect the incidence of AF in patients with a cryptogenic stroke or transient ischemic attack (TIA) and to evaluate the association between the presence of AF and selected single-nucleotide polymorphisms (SNP).
Methods: Patients with a cryptogenic stroke/ TIA (n=100) and a control group (n=15) of volunteers without significant cardiovascular disease were included in the study during the period of 2014 to 2019. To detect AF they underwent 12 months of ECG monitoring using an implanted loop recorder (ILR). Genotyping for SNPs rs10033464, rs2200733, rs225132, and rs2106261 was performed by a high resolution melting analysis.
Results: We found AF to be present in 24 (24%) patients with a cryptogenic stroke/TIA, versus no subjects in the control group. The SNPs rs2106261, rs2200733, rs225132, and rs10033464 were not found to be associated with AF in our study (p=0.240; 1.000; 0.887; 0.589). However, a weak trend for a higher frequency of rs2106261 risk allele A homozygotes was observed in the patients with AF compared to the patients without AF (0.416 vs. 0.263, p=0.073). Homozygotes for allele A of rs2106261 were also present in a significantly higher frequency in AF patients compared to the controls (0.416 vs. 0.133, p = 0.012).
Conclusion: In our study paroxysmal AF was a probable etiological factor in 24% of patients with cryptogenic ischemic stroke / TIA during the 12 months of monitoring. The homozygous allele A of rs2106261 was identified to be the possible genetic risk factor of AF, but this should be verified in larger cohorts.
The study has been registered at www.clinicaltrials.gov, identifier NCT02216370.
Collapse
|
35
|
Kornej J, Hanger VA, Trinquart L, Ko D, Preis SR, Benjamin EJ, Lin H. New biomarkers from multiomics approaches: improving risk prediction of atrial fibrillation. Cardiovasc Res 2021; 117:1632-1644. [PMID: 33751041 PMCID: PMC8208748 DOI: 10.1093/cvr/cvab073] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/07/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
Atrial fibrillation (AF) is a common cardiac arrhythmia leading to many adverse outcomes and increased mortality. Yet the molecular mechanisms underlying AF remain largely unknown. Recent advances in high-throughput technologies make large-scale molecular profiling possible. In the past decade, multiomics studies of AF have identified a number of potential biomarkers of AF. In this review, we focus on the studies of multiomics profiles with AF risk. We summarize recent advances in the discovery of novel biomarkers for AF through multiomics studies. We also discuss limitations and future directions in risk assessment and discovery of therapeutic targets for AF.
Collapse
Affiliation(s)
- Jelena Kornej
- National Heart, Lung, and Blood Institute’s Framingham Heart Study, 73 Mt Wayte Ave, Framingham, MA 01702, USA
- Section of Cardiovascular Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | | | - Ludovic Trinquart
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Darae Ko
- Section of Cardiovascular Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Sarah R Preis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Emelia J Benjamin
- National Heart, Lung, and Blood Institute’s Framingham Heart Study, 73 Mt Wayte Ave, Framingham, MA 01702, USA
- Section of Cardiovascular Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Section of Preventive Medicine & Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Honghuang Lin
- National Heart, Lung, and Blood Institute’s Framingham Heart Study, 73 Mt Wayte Ave, Framingham, MA 01702, USA
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
36
|
Gharanei M, Shafaattalab S, Sangha S, Gunawan M, Laksman Z, Hove-Madsen L, Tibbits GF. Atrial-specific hiPSC-derived cardiomyocytes in drug discovery and disease modeling. Methods 2021; 203:364-377. [PMID: 34144175 DOI: 10.1016/j.ymeth.2021.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/08/2021] [Accepted: 06/12/2021] [Indexed: 12/19/2022] Open
Abstract
The discovery and application of human-induced pluripotent stem cells (hiPSCs) have been instrumental in the investigation of the pathophysiology of cardiovascular diseases. Patient-specific hiPSCs can now be generated, genome-edited, and subsequently differentiated into various cell types and used for regenerative medicine, disease modeling, drug testing, toxicity screening, and 3D tissue generation. Modulation of the retinoic acid signaling pathway has been shown to direct cardiomyocyte differentiation towards an atrial lineage. A variety of studies have successfully differentiated patient-specific atrial cardiac myocytes (hiPSC-aCM) and atrial engineered heart tissue (aEHT) that express atrial specific genes (e.g., sarcolipin and ANP) and exhibit atrial electrophysiological and contractility profiles. Identification of protocols to differentiate atrial cells from patients with atrial fibrillation and other inherited diseases or creating disease models using genetic mutation studies has shed light on the mechanisms of atrial-specific diseases and identified the efficacy of atrial-selective pharmacological compounds. hiPSC-aCMs and aEHTs can be used in drug discovery and drug screening studies to investigate the efficacy of atrial selective drugs on atrial fibrillation models. Furthermore, hiPSC-aCMs can be effective tools in studying the mechanism, pathophysiology and treatment options of atrial fibrillation and its genetic underpinnings. The main limitation of using hiPSC-CMs is their immature phenotype compared to adult CMs. A wide range of approaches and protocols are used by various laboratories to optimize and enhance CM maturation, including electrical stimulation, culture time, biophysical cues and changes in metabolic factors.
Collapse
Affiliation(s)
- Mayel Gharanei
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada; hiPSC-CM Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Sanam Shafaattalab
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada; hiPSC-CM Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Sarabjit Sangha
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada; hiPSC-CM Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Marvin Gunawan
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada; hiPSC-CM Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Zachary Laksman
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Leif Hove-Madsen
- Cardiac Rhythm and Contraction Group, IIBB-CSIC, CIBERCV, IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona 08025, Spain
| | - Glen F Tibbits
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada; hiPSC-CM Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
37
|
Thorolfsdottir RB, Sveinbjornsson G, Aegisdottir HM, Benonisdottir S, Stefansdottir L, Ivarsdottir EV, Halldorsson GH, Sigurdsson JK, Torp-Pedersen C, Weeke PE, Brunak S, Westergaard D, Pedersen OB, Sorensen E, Nielsen KR, Burgdorf KS, Banasik K, Brumpton B, Zhou W, Oddsson A, Tragante V, Hjorleifsson KE, Davidsson OB, Rajamani S, Jonsson S, Torfason B, Valgardsson AS, Thorgeirsson G, Frigge ML, Thorleifsson G, Norddahl GL, Helgadottir A, Gretarsdottir S, Sulem P, Jonsdottir I, Willer CJ, Hveem K, Bundgaard H, Ullum H, Arnar DO, Thorsteinsdottir U, Gudbjartsson DF, Holm H, Stefansson K. Genetic insight into sick sinus syndrome. Eur Heart J 2021; 42:1959-1971. [PMID: 36282123 PMCID: PMC8140484 DOI: 10.1093/eurheartj/ehaa1108] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/24/2020] [Accepted: 01/05/2021] [Indexed: 12/19/2022] Open
Abstract
Aims The aim of this study was to use human genetics to investigate the pathogenesis of sick sinus syndrome (SSS) and the role of risk factors in its development. Methods and results We performed a genome-wide association study of 6469 SSS cases and 1 000 187 controls from deCODE genetics, the Copenhagen Hospital Biobank, UK Biobank, and the HUNT study. Variants at six loci associated with SSS, a reported missense variant in MYH6, known atrial fibrillation (AF)/electrocardiogram variants at PITX2, ZFHX3, TTN/CCDC141, and SCN10A and a low-frequency (MAF = 1.1–1.8%) missense variant, p.Gly62Cys in KRT8 encoding the intermediate filament protein keratin 8. A full genotypic model best described the p.Gly62Cys association (P = 1.6 × 10−20), with an odds ratio (OR) of 1.44 for heterozygotes and a disproportionally large OR of 13.99 for homozygotes. All the SSS variants increased the risk of pacemaker implantation. Their association with AF varied and p.Gly62Cys was the only variant not associating with any other arrhythmia or cardiovascular disease. We tested 17 exposure phenotypes in polygenic score (PGS) and Mendelian randomization analyses. Only two associated with the risk of SSS in Mendelian randomization, AF, and lower heart rate, suggesting causality. Powerful PGS analyses provided convincing evidence against causal associations for body mass index, cholesterol, triglycerides, and type 2 diabetes (P > 0.05). Conclusion We report the associations of variants at six loci with SSS, including a missense variant in KRT8 that confers high risk in homozygotes and points to a mechanism specific to SSS development. Mendelian randomization supports a causal role for AF in the development of SSS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jon K Sigurdsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | - Christian Torp-Pedersen
- Department of Clinical Research and Cardiology, Nordsjaelland Hospital, Dyrehavevej 29, Hillerød 3400, Denmark
| | - Peter E Weeke
- Department of Cardiology, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen 2100, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, Copenhagen 2200, Denmark
| | - David Westergaard
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, Copenhagen 2200, Denmark
| | - Ole B Pedersen
- Department of Clinical Immunology, Naestved Hospital, Ringstedgade 77B, Naestved 4700, Denmark
| | - Erik Sorensen
- Department of Clinical Immunology, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen 2100, Denmark
| | - Kaspar R Nielsen
- Department of Clinical Immunology, Aalborg University Hospital North, Urbansgade 36, Aalborg 9000, Denmark
| | - Kristoffer S Burgdorf
- Department of Clinical Immunology, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen 2100, Denmark
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, Copenhagen 2200, Denmark
| | - Ben Brumpton
- Department of Thoracic and Occupational Medicine, St. Olavs Hospital, Trondheim University Hospital, Prinsesse Kristinas gate 3, Trondheim 7030, Norway
| | - Wei Zhou
- Department of Computational Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Avenue, Ann Arbor, MI 48109-2218, USA
| | - Asmundur Oddsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | | | - Kristjan E Hjorleifsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Department of Computing and Mathematical Sciences, California Institute of Technology, 1200 E California Blvd. MC 305-16, Pasadena, CA 91125, USA
| | | | | | - Stefan Jonsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | - Bjarni Torfason
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland.,Department of Cardiothoracic Surgery, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Atli S Valgardsson
- Department of Cardiothoracic Surgery, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Gudmundur Thorgeirsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland.,Department of Medicine, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Michael L Frigge
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | | | | | - Anna Helgadottir
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | | | - Patrick Sulem
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | - Ingileif Jonsdottir
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland.,Department of Immunology, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Cristen J Willer
- Department of Computational Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Avenue, Ann Arbor, MI 48109-2218, USA.,Department of Internal Medicine: Cardiology, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109 -5368, USA.,Department of Human Genetics, University of Michigan, 4909 Buhl Building, 1241 E. Catherine St., Ann Arbor, MI 48109 -5618, USA
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Erling Skjalgssons gt. 1, Trondheim 7491, Norway.,Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Postboks 8905, Trondheim 7491, Norway.,HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Forskningsveien 2, Levanger 7600, Norway
| | - Henning Bundgaard
- Department of Cardiology, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen 2100, Denmark
| | - Henrik Ullum
- Department of Clinical Immunology, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen 2100, Denmark.,Statens Serum Institut, Artillerivej 5, Copenhagen 2300, Denmark
| | - David O Arnar
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland.,Department of Medicine, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,School of Engineering and Natural Sciences, University of Iceland, Hjardarhagi 4, Reykjavik 107, Iceland
| | - Hilma Holm
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | - Kari Stefansson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland
| | | |
Collapse
|
38
|
Dai W, Kesaraju S, Weber CR. Transcriptional factors in calcium mishandling and atrial fibrillation development. Pflugers Arch 2021; 473:1177-1197. [PMID: 34003377 DOI: 10.1007/s00424-021-02553-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/19/2021] [Accepted: 02/05/2021] [Indexed: 12/19/2022]
Abstract
Healthy cardiac conduction relies on the coordinated electrical activity of distinct populations of cardiomyocytes. Disruption of cell-cell conduction results in cardiac arrhythmias, a leading cause of morbidity and mortality worldwide. Recent genetic studies have highlighted a major heritable component and identified numerous loci associated with risk of atrial fibrillation, including transcription factor genes, particularly those important in cardiac development, microRNAs, and long noncoding RNAs. Identification of such genetic factors has prompted the search to understand the mechanisms that underlie the genetic component of AF. Recent studies have found several mechanisms by which genetic alterations can result in AF formation via disruption of calcium handling. Loss of developmental transcription factors in adult cardiomyocytes can result in disruption of SR calcium ATPase, sodium calcium exchanger, calcium channels, among other ion channels, which underlie action potential abnormalities and triggered activity that can contribute to AF. This review aims to summarize the complex network of transcription factors and their roles in calcium handling.
Collapse
Affiliation(s)
- Wenli Dai
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Sneha Kesaraju
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | | |
Collapse
|
39
|
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: 7] [Impact Index Per Article: 1.8] [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
|
40
|
Victorino J, Alvarez-Franco A, Manzanares M. Functional genomics and epigenomics of atrial fibrillation. J Mol Cell Cardiol 2021; 157:45-55. [PMID: 33887329 DOI: 10.1016/j.yjmcc.2021.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023]
Abstract
Atrial fibrillation is a progressive cardiac arrhythmia that increases the risk of hospitalization and adverse cardiovascular events. Despite years of study, we still do not have a full comprehension of the molecular mechanism responsible for the disease. The recent implementation of large-scale approaches in both patient samples, population studies and animal models has helped us to broaden our knowledge on the molecular drivers responsible for AF and on the mechanisms behind disease progression. Understanding genomic and epigenomic changes that take place during chronification of AF will prove essential to design novel treatments leading to improved patient care.
Collapse
Affiliation(s)
- Jesus Victorino
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Spain
| | - Alba Alvarez-Franco
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Miguel Manzanares
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.
| |
Collapse
|
41
|
Rienstra M, Siland JE, Ellinor PT. Role of genetics in atrial fibrillation management. Europace 2021; 23:ii4-ii8. [PMID: 33837754 PMCID: PMC8035706 DOI: 10.1093/europace/euaa366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/13/2020] [Indexed: 01/17/2023] Open
Abstract
Atrial fibrillation (AF) management has significantly improved during the career of professor Crijns. Research was implemented into guidelines and clinical practice. However, despite advances in AF management, large differences between individual treatment responses still exist and the mechanisms underlying initiation and perpetuation of AF are not completely understood. International collaborations have revealed the genetic contribution to AF and steps towards improving AF management are being made. In this short review, the most important paradigms shifts in the field of AF genetics are recognized and the future role of genetics in personalized management of AF is discussed.
Collapse
Affiliation(s)
- Michiel Rienstra
- Department of Cardiology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Joylene E Siland
- Department of Cardiology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiac Arrhythmia Service, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
42
|
Lkhagva B, Lin Y, Chen Y, Cheng W, Higa S, Kao Y, Chen Y. ZFHX3 knockdown dysregulates mitochondrial adaptations to tachypacing in atrial myocytes through enhanced oxidative stress and calcium overload. Acta Physiol (Oxf) 2021; 231:e13604. [PMID: 33332716 DOI: 10.1111/apha.13604] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 12/06/2020] [Accepted: 12/11/2020] [Indexed: 01/02/2023]
Abstract
AIM To investigate the role of zinc finger homeobox 3 gene (ZFHX3) in tachypacing-induced mitochondrial dysfunction and explore its molecular mechanisms and potential as a therapeutic target in atrial fibrillation (AF). METHODS Through a bioluminescent assay, a patch clamp, confocal fluorescence and fluorescence microscopy, microplate enzyme activity assays and Western blotting, we studied ATP and ADP production, mitochondrial electron transfer chain complex activities, ATP-sensitive potassium channels (IKATP ), mitochondrial oxidative stress, Ca2+ content, and protein expression in control and ZFHX3 knockdown (KD) HL-1 cells subjected to 1 and 5-Hz pacing for 24 hours. RESULTS Compared with 1-Hz pacing, 5-Hz pacing increased ATP and ADP production, IKATP , phosphorylated adenosine monophosphate-activated protein kinase and inositol 1,4,5-triphosphate (IP3 ) receptor (IP3 R) protein expression. Tachypacing induced mitochondrial oxidative stress and Ca2+ overload in both cell types. Furthermore, under 1- and 5-Hz pacing, ZFHX3 KD cells showed higher IKATP , ATP and ADP production, mitochondrial oxidative stress and Ca2+ content than control cells. Under 5-Hz pacing, 2-aminoethoxydiphenyl borate (2-APB; 3 μmol/L, an IP3 R inhibitor) and MitoTEMPO (10 µmol/L, a mitochondria-targeted antioxidant) reduced ADP and increased ATP production in both cell types; however, only 2-APB significantly reduced mitochondrial Ca2+ overload in control cells. Under 5-Hz pacing, mitochondrial oxidative stress was significantly reduced by both MitoTEMPO and 2-APB and only by 2-APB in control and ZFHX3 KD cells respectively. CONCLUSION ZFHX3 KD cells modulate mitochondrial adaptations to tachypacing in HL-1 cardiomyocytes through Ca2+ overload, oxidative stress and metabolic disorder. Targeting IP3 R signalling or oxidative stress could reduce AF.
Collapse
Affiliation(s)
- Baigalmaa Lkhagva
- Graduate Institute of Clinical Medicine College of Medicine Taipei Medical University Taipei Taiwan
| | - Yung‐Kuo Lin
- Division of Cardiology Department of Internal Medicine School of Medicine College of Medicine Taipei Medical University Taipei Taiwan
| | - Yao‐Chang Chen
- Department of Biomedical Engineering National Defense Medical Center Taipei Taiwan
| | - Wan‐Li Cheng
- Graduate Institute of Clinical Medicine College of Medicine Taipei Medical University Taipei Taiwan
| | - Satoshi Higa
- Cardiac Electrophysiology and Pacing Laboratory Division of Cardiovascular Medicine Makiminato Central Hospital Okinawa Japan
| | - Yu‐Hsun Kao
- Graduate Institute of Clinical Medicine College of Medicine Taipei Medical University Taipei Taiwan
- Department of Medical Education and Research Wan‐Fang Hospital Taipei Medical University Taipei Taiwan
| | - Yi‐Jen Chen
- Graduate Institute of Clinical Medicine College of Medicine Taipei Medical University Taipei Taiwan
- Cardiovascular Research CenterWan‐Fang HospitalTaipei Medical University Taipei Taiwan
| |
Collapse
|
43
|
Bai J, Zhu Y, Lo A, Gao M, Lu Y, Zhao J, Zhang H. In Silico Assessment of Class I Antiarrhythmic Drug Effects on Pitx2-Induced Atrial Fibrillation: Insights from Populations of Electrophysiological Models of Human Atrial Cells and Tissues. Int J Mol Sci 2021; 22:1265. [PMID: 33514068 PMCID: PMC7866025 DOI: 10.3390/ijms22031265] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
Electrical remodelling as a result of homeodomain transcription factor 2 (Pitx2)-dependent gene regulation was linked to atrial fibrillation (AF) and AF patients with single nucleotide polymorphisms at chromosome 4q25 responded favorably to class I antiarrhythmic drugs (AADs). The possible reasons behind this remain elusive. The purpose of this study was to assess the efficacy of the AADs disopyramide, quinidine, and propafenone on human atrial arrhythmias mediated by Pitx2-induced remodelling, from a single cell to the tissue level, using drug binding models with multi-channel pharmacology. Experimentally calibrated populations of human atrial action po-tential (AP) models in both sinus rhythm (SR) and Pitx2-induced AF conditions were constructed by using two distinct models to represent morphological subtypes of AP. Multi-channel pharmaco-logical effects of disopyramide, quinidine, and propafenone on ionic currents were considered. Simulated results showed that Pitx2-induced remodelling increased maximum upstroke velocity (dVdtmax), and decreased AP duration (APD), conduction velocity (CV), and wavelength (WL). At the concentrations tested in this study, these AADs decreased dVdtmax and CV and prolonged APD in the setting of Pitx2-induced AF. Our findings of alterations in WL indicated that disopyramide may be more effective against Pitx2-induced AF than propafenone and quinidine by prolonging WL.
Collapse
Affiliation(s)
- Jieyun Bai
- Department of Electronic Engineering, College of Information Science and Technology, Jinan University, Guangzhou 510632, China;
| | - Yijie Zhu
- Department of Electronic Engineering, College of Information Science and Technology, Jinan University, Guangzhou 510632, China;
| | - Andy Lo
- Auckland Bioengineering Institute, University of Auckland, Auckland 1010, New Zealand; (A.L.); (J.Z.)
| | - Meng Gao
- Department of Computer Science and Technology, College of Electrical Engineering and Information, Northeast Agricultural University, Harbin 150030, China
| | - Yaosheng Lu
- Department of Electronic Engineering, College of Information Science and Technology, Jinan University, Guangzhou 510632, China;
| | - Jichao Zhao
- Auckland Bioengineering Institute, University of Auckland, Auckland 1010, New Zealand; (A.L.); (J.Z.)
| | - Henggui Zhang
- Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester M13 9PL, UK;
| |
Collapse
|
44
|
Zhang J, Johnsen SP, Guo Y, Lip GYH. Epidemiology of Atrial Fibrillation: Geographic/Ecological Risk Factors, Age, Sex, Genetics. Card Electrophysiol Clin 2021; 13:1-23. [PMID: 33516388 DOI: 10.1016/j.ccep.2020.10.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Atrial fibrillation is the most common arrhythmia globally. The global prevalence of atrial fibrillation is positively correlated with the sociodemographic index of different regions. Advancing age, male sex, and Caucasian race are risk factors; female sex is correlated with higher atrial fibrillation mortality worldwide likely owing to thromboembolic risk. African American ethnicity is associated with lower atrial fibrillation risk, same as Asian and Hispanic/Latino ethnicities compared with Caucasians. Atrial fibrillation may be heritable, and more than 100 genetic loci have been identified. A polygenic risk score and clinical risk factors are feasible and effective in risk stratification of incident disease.
Collapse
Affiliation(s)
- Juqian Zhang
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, L14 3PE, UK
| | - Søren Paaske Johnsen
- Department of Clinical Medicine, Aalborg University, Søndre Skovvej 15, Aalborg, Aalborg 9000, Denmark
| | - Yutao Guo
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, L14 3PE, UK; Department of Cardiology, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, L14 3PE, UK; Department of Clinical Medicine, Aalborg University, Søndre Skovvej 15, Aalborg, Aalborg 9000, Denmark; Department of Cardiology, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
45
|
Park YM, Roh SY, Lee DI, Shim J, Choi JI, Park SW, Kim YH. The Effects of Single Nucleotide Polymorphisms in Korean Patients with Early-onset Atrial Fibrillation after Catheter Ablation. J Korean Med Sci 2020; 35:e411. [PMID: 33350184 PMCID: PMC7752257 DOI: 10.3346/jkms.2020.35.e411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/13/2020] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND This study evaluated the status of single nucleotide polymorphisms (SNPs) in Korean patients with early-onset (< 40 years old) atrial fibrillation (AF) and their effects on the outcome after catheter ablation. METHODS A total of 89 patients (35.7 ± 3.7 years, 81 males) with drug-refractory AF (paroxysmal 64.0%) who underwent catheter ablation were included in this study. Sixteen SNPs, including rs13376333, rs10465885, rs10033464, rs2200733, rs17042171, rs6843082, rs7193343, rs2106261, rs17570669, rs853445, rs11708996, rs6800541, rs251253, rs3807989, rs11047543, and rs3825214, were genotyped. Serial 48-hour Holter monitoring was conducted to detect AF recurrences during long-term follow up. RESULTS Wild-type genotypes of rs11047543 (GG; 26/69 [37.7%] vs. GA; 13/18 [72.2%] vs. AA; 0/0 [0%], P = 0.009) and rs7193343 (CC; 0/7 [0%] vs. CT; 22/40 [55.0%] vs. TT; 18/41 [43.9%], P = 0.025) and the homozygous variant of rs3825214 (AA; 16/31 [51.6%] vs. AG; 22/43 [51.2%] vs. GG; 2/13 [15.4%], P = 0.056) were significantly associated with a lower rate of late recurrence. When the patients were assigned to four groups according to the number of risk alleles (n = 0-3), there were significant differences in recurrence rate (n = 0; 0/3 vs. n = 1; 2/13 [15.4%] vs. n = 2; 24/52 [46.2%] vs. n = 3; 13/17 [76.5%], P = 0.003). When correcting for multiple variables, rs11047543 (hazard ratio [HR], 2.723; 95% confidence interval [CI], 1.358-5.461; P = 0.005) and the number of risk alleles (HR, 2.901; 95% CI, 1.612-5.219; P < 0.001) were significantly associated with recurrence of AF after catheter ablation. CONCLUSION Polymorphisms on rs7193343 closest to ZFHX3 (16q22), rs3825214 near to TBX5 (12q24), and rs11047543 near to SOX5 (12p12) modulate the risk for AF recurrence after catheter ablation. The number of risk alleles of these 3 SNPs was an independent predictor of recurrence during long-term follow up in Korean patients with early-onset AF.
Collapse
Affiliation(s)
- Yae Min Park
- Division of Cardiology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Korea
| | - Seung Young Roh
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Dae In Lee
- Division of Cardiology, Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Korea
| | - Jaemin Shim
- Division of Cardiology, Department of Internal Medicine, Korea University Anam Hospital, Seoul, Korea
| | - Jong Il Choi
- Division of Cardiology, Department of Internal Medicine, Korea University Anam Hospital, Seoul, Korea
| | - Sang Weon Park
- Division of Cardiology, Department of Internal Medicine, Sejong General Hospital, Bucheon, Korea
| | - Young Hoon Kim
- Division of Cardiology, Department of Internal Medicine, Korea University Anam Hospital, Seoul, Korea.
| |
Collapse
|
46
|
Atrial fibrillation-a complex polygenetic disease. Eur J Hum Genet 2020; 29:1051-1060. [PMID: 33279945 PMCID: PMC8298566 DOI: 10.1038/s41431-020-00784-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 10/27/2020] [Accepted: 11/17/2020] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most common type of arrhythmia. Epidemiological studies have documented a substantial genetic component. More than 160 genes have been associated with AF during the last decades. Some of these were discovered by classical linkage studies while the majority relies on functional studies or genome-wide association studies. In this review, we will evaluate the genetic basis of AF and the role of both common and rare genetic variants in AF. Rare variants in multiple ion-channel genes as well as gap junction and transcription factor genes have been associated with AF. More recently, a growing body of evidence has implicated structural genes with AF. An increased burden of atrial fibrosis in AF patients compared with non-AF patients has also been reported. These findings challenge our traditional understanding of AF being an electrical disease. We will focus on several quantitative landmark papers, which are transforming our understanding of AF by implicating atrial cardiomyopathies in the pathogenesis. This new AF research field may enable better diagnostics and treatment in the future.
Collapse
|
47
|
Johansson C, Lind MM, Eriksson M, Johansson L. Weight, height, weight change, and risk of incident atrial fibrillation in middle-aged men and women. J Arrhythm 2020; 36:974-981. [PMID: 33335612 PMCID: PMC7733566 DOI: 10.1002/joa3.12409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/18/2020] [Accepted: 06/28/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Anthropometric factors are reported to be risk factors for atrial fibrillation (AF), but it is unclear whether weight change in mid-life is associated with AF. We aimed to study the possible associations of weight, height, and weight change with the risk of incident AF in men and women. METHODS Our study cohort included 108 417 persons (51% women) who participated in a population-based health examination in northern Sweden at 30, 40, 50, or 60 years of age. The health examination included weight and height measurement and collection of data regarding cardiovascular risk factors. Within this cohort, 40 275 participants underwent two health examinations with a 10-year interval. We identified cases with a first-ever diagnosis of AF through the Swedish National Patient Registry. RESULTS During a total follow-up of 1 469 820 person-years, 5154 participants developed incident AF. The mean age at inclusion was 46.3 years, and mean age at AF diagnosis was 66.6 years. After adjustment for potential confounders, height, weight, body mass index (BMI), and body surface area (BSA) were positively associated with risk of incident AF in both men and women. Among participants who underwent two health examinations 10 years apart, 1142 persons developed AF. The mean weight change from baseline was a gain of 4.8%. Weight gain or weight loss was not significantly associated with risk of incident AF. CONCLUSIONS Height, weight, BMI, and BSA showed positive associations with risk of incident AF in both men and women. Midlife weight change was not significantly associated with AF risk.
Collapse
Affiliation(s)
- Cecilia Johansson
- Skellefteå Research UnitDepartment of Public Health and Clinical MedicineUmeå UniversitySkellefteåSweden
| | - Marcus M. Lind
- Skellefteå Research UnitDepartment of Public Health and Clinical MedicineUmeå UniversitySkellefteåSweden
| | | | - Lars Johansson
- Skellefteå Research UnitDepartment of Public Health and Clinical MedicineUmeå UniversitySkellefteåSweden
| |
Collapse
|
48
|
Li R, Chen W, Mao P, Wang J, Jing J, Sun Q, Wang M, Yu X. Identification of a three-long non-coding RNA signature for predicting survival of temozolomide-treated isocitrate dehydrogenase mutant low-grade gliomas. Exp Biol Med (Maywood) 2020; 246:187-196. [PMID: 33028081 DOI: 10.1177/1535370220962715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Temozolomide (TMZ) is the major chemotherapy agent in glioma, and isocitrate dehydrogenase (IDH) is a well-known prognostic marker in glioma. O6-methylguanine-DNA methyltransferase promoter methylation (MGMTmethyl) is a predictive biomarker in overall gliomas rather than in IDH mutant gliomas. To discover effective biomarkers that could predict TMZ efficacy in IDH mutant low-grade gliomas (LGGs), we retrieved data of IDH mutant LGGs from TMZ arm of the EORTC22033-26033 trial as the training-set (n = 83), analyzed correlations between long non-coding RNAs (lncRNAs) and progression-free survival (PFS) using Lasso-Cox regression, and created a risk score (RS) to stratify patients. We identified a three-lncRNA signature in TMZ-treated IDH mutant LGGs. All of the three lncRNAs, as well as the RS derived, were significantly correlated with PFS. Patients were classified into high-risk and low-risk groups according to RS. PFS of the high-risk group was significantly worse than that of the low-risk group (P < 0.001). AUCs of the three-, four-, and five-year survival probability predicted by RS were 0.73, 0.79, and 0.76, respectively. The predictive role of the three-lncRNA signature was further validated in an independent testing-set, the TCGA-LGGs, which resulted in a significantly worse PFS (P < 0.001) in the high-risk group. Three-, four-, and five-year survival probabilities predicted by RS were 0.65, 0.69, and 0.84, respectively. Functions of these three lncRNAs involve cell proliferation and differentiation, predicted by their targeting cancer genes. Conclusively, we created a scoring model based on the expression of three lncRNAs, which can effectively predict the survival of IDH mutant LGGs treated with TMZ.
Collapse
Affiliation(s)
- Ruichun Li
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Wei Chen
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Ping Mao
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jia Wang
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jiangpeng Jing
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Qinli Sun
- Department of Diagnostic Radiology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Maode Wang
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiao Yu
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
49
|
Tidbury N, Preston J, Ding WY, Rivera-Caravaca JM, Marín F, Lip GYH. Utilizing biomarkers associated with cardiovascular events in atrial fibrillation: informing a precision medicine response. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020. [DOI: 10.1080/23808993.2020.1804864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Nicola Tidbury
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
| | - Joshua Preston
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
| | - Wern Yew Ding
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
| | - José Miguel Rivera-Caravaca
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
- Department of Cardiology, Hospital Clínico Universitario Virgen De La Arrixaca, University of Murcia, Instituto Murciano De Investigación Biosanitaria (Imib-arrixaca), CIBERCV, Murcia, Spain
| | - Francisco Marín
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
- Department of Cardiology, Hospital Clínico Universitario Virgen De La Arrixaca, University of Murcia, Instituto Murciano De Investigación Biosanitaria (Imib-arrixaca), CIBERCV, Murcia, Spain
| | - Gregory Y. H. Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
- Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
50
|
Xu C, Zhang R, Xia Y, Xiong L, Yang W, Wang P. Annotation of susceptibility SNPs associated with atrial fibrillation. Aging (Albany NY) 2020; 12:16981-16998. [PMID: 32902410 PMCID: PMC7521544 DOI: 10.18632/aging.103615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 06/18/2020] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Genome-wide association studies (GWAS) and the candidate gene based association studies have identified a panel of variants associated with atrial fibrillation (AF), however, most of the identified single nucleotide polymorphisms (SNPs) were found located within intergenic or intronic genomic regions, and whether the positive SNPs have a real biological function is unknown, and the real disease causing gene need to be studied. RESULTS The current results of the genetic studies including common variants identified by GWAS (338 index SNPs) and candidate gene based association studies (40 SNPs) were summarized. CONCLUSION Our study suggests the relationship between genetic variants and possible targeted genes, and provides insight into potential genetic pathways underlying AF incidence and development. The results may provide an encyclopedia of AF susceptibility SNPs and shed light on the functional mechanisms of AF variants identified through genetic studies. METHODS We summarized AF susceptibility SNPs identified by GWAS and candidate gene based association studies, and give a comprehensive functional annotation of all these AF susceptibility loci. by genomic annotation, microRNA binding prediction, promoter activity analysis, enhancer activity analysis, transcription factors binding activity prediction, expression quantitative trait loci (eQTL) analysis, long-range transcriptional regulatory function analysis, gene ontology and pathway enrichment analysis.
Collapse
Affiliation(s)
- Chengqi Xu
- College of Life Science and Technology, Center for Human Genome Research and Cardio-X Institute, Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Rongfeng Zhang
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian 116011, P. R. China
| | - Yunlong Xia
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian 116011, P. R. China
| | - Liang Xiong
- Department of Clinical Laboratory, Liyuan Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, P. R. China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Pengyun Wang
- Department of Clinical Laboratory, Liyuan Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, P. R. China
| |
Collapse
|