1
|
Zhao Y, Wan J, Liao B, Qi M. The Neglected Internodal Tract-A Cardiac Conduction System Structure Homologous to the Development and Regulation of the Sinoatrial Node. Rev Cardiovasc Med 2025; 26:27882. [PMID: 40351691 PMCID: PMC12059794 DOI: 10.31083/rcm27882] [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: 11/04/2024] [Revised: 12/05/2024] [Accepted: 12/12/2024] [Indexed: 05/14/2025] Open
Abstract
The existence of internodal tracts (ITs) is controversial. Indeed, ITs in the cardiac conduction system (CCS), connected to the sinoatrial node (SAN), transmit electrical signals quickly to the left atrium and the atrioventricular node (AVN). Interestingly, research has suggested that the ITs and the tail of the SAN may share developmental homology. Additionally, many studies indicate that ITs blockage can lead to atrial conduction block and is associated with atrial fibrillation (AF). However, few studies have been reported on the morphogenesis, development, and function of ITs. Therefore, this paper aims to review the morphogenesis, development, and function of ITs, focusing on the regulatory mechanisms of transcription factors (TFs), such as NK2 homeobox 5 (NKX2.5), SHOX homeobox 2 (SHOX2), hyperpolarization activated cyclic nucleotide gated potassium channel 4 (HCN4), and T-box transcription factor 3 (TBX3) in the development and morphogenesis of ITs. This review also explores the causes of arrhythmias, especially atrial block, in order to provide new insights into the pathogenesis of CCS disorders.
Collapse
Affiliation(s)
- Yuanqin Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
- Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, 646000 Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Juyi Wan
- Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, 646000 Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, 646000 Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Man Qi
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
- Chinese People’s Liberation Army (PLA) General Hospital, College of Pulmonary & Critical Care Medicine, 100091 Beijing, China
- Department of Cardiology, Chinese PLA General Hospital, 100853 Beijing, China
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, 200120 Shanghai, China
| |
Collapse
|
2
|
Yan Z, Pu X, Chang X, Liu Z, Liu R. Genetic basis and causal relationship between atrial fibrillation and sinus node dysfunction: Evidence from comprehensive genetic analysis. Int J Cardiol 2025; 418:132609. [PMID: 39389108 DOI: 10.1016/j.ijcard.2024.132609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/16/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Atrial fibrillation (AF) and sinus node dysfunction (SND) are commonly observed together clinically. However, little is known about the genetic background and causal relationship between the two. METHODS Firstly, we investigated the global and local genetic correlations between AF and SND using LDSC and HESS. Then, we identified shared "Novel SNPs" between AF and SND through two complementary cross-trait meta-analyses and mapped the "pleiotropic genes" behind these SNPs, validated by colocalization analysis. Additionally, we explored the degree of genetic enrichment of SNPs in specific tissues using LDSC-SEG and MAGMA, and identified potential functional genes in tissues using summary data-based Mendelian randomization (SMR). Finally, two-sample Mendelian randomization (TSMR) and multivariable Mendelian randomization (MVMR) were used to explore the causal relationship between AF and SND. RESULTS Both global and local genetic correlation analyses revealed a high positive genetic correlation between AF and SND. HESS identified 9 shared loci, with chr4(q25-q26) and chr11(p11.12-q11) being prominent. Cross-trait meta-analysis and colocalization analysis identified ENPEP and PITX2 as novel pleiotropic genes. MAGMA revealed genetic enrichment of SNPs for AF and SND in the "Heart Left Ventricle" and "Heart Atrial Appendage" tissues, with CEP68 and BEST3 identified as potential functional genes through SMR. MR analysis indicated that AF increases the risk of SND, even after adjusting for confounding factors. CONCLUSION This study provides genetic evidence for the increased risk of SND associated with AF, identifying multiple shared risk loci and enriched tissues, and discovering 2 novel pleiotropic genes and 2 new functional genes.
Collapse
Affiliation(s)
- Zhaoqi Yan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiangyi Pu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Heriot-Watt University, Edinburgh, United Kingdom
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhiming Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruxiu Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
3
|
Li H, Tang Q, Yang T, Wang Z, Li D, Wang L, Li L, Chen Y, Huang H, Zhang Y, Chen Y. Segregation of morphogenetic regulatory function of Shox2 from its cell fate guardian role in sinoatrial node development. Commun Biol 2024; 7:385. [PMID: 38553636 PMCID: PMC10980793 DOI: 10.1038/s42003-024-06039-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 03/11/2024] [Indexed: 04/02/2024] Open
Abstract
Shox2 plays a vital role in the morphogenesis and physiological function of the sinoatrial node (SAN), the primary cardiac pacemaker, manifested by the formation of a hypoplastic SAN and failed differentiation of pacemaker cells in Shox2 mutants. Shox2 and Nkx2-5 are co-expressed in the developing SAN and regulate the fate of the pacemaker cells through a Shox2-Nkx2-5 antagonistic mechanism. Here we show that simultaneous inactivation of Nkx2-5 in the SAN of Shox2 mutants (dKO) rescued the pacemaking cell fate but not the hypoplastic defects, indicating uncoupling of SAN cell fate determination and morphogenesis. Single-cell RNA-seq revealed that the presumptive SAN cells of Shox2-/- mutants failed to activate pacemaking program but remained in a progenitor state preceding working myocardium, while both wildtype and dKO SAN cells displayed normal pacemaking cell fate with similar cellular state. Shox2 thus acts as a safeguard but not a determinant to ensure the pacemaking cell fate through the Shox2-Nkx2-5 antagonistic mechanism, which is segregated from its morphogenetic regulatory function in SAN development.
Collapse
Affiliation(s)
- Hua Li
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China.
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA.
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province, 350108, PR China.
| | - Qinghuang Tang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, 14214, USA
| | - Tianfang Yang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Zhengsen Wang
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province, 350108, PR China
| | - Dainan Li
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Linyan Wang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
- Department of Stomatology, Chengdu Second People's Hospital, Chengdu, Sichuan Province, 610021, PR China
| | - Liwen Li
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
- Department of Biological Sciences, College of Arts and Sciences, University at Buffalo, Buffalo, NY, 14260, USA
| | - Yaoyi Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Hai Huang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Yanding Zhang
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province, 350108, PR China
| | - YiPing Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA.
| |
Collapse
|
4
|
Lee C, Xu S, Samad T, Goodyer WR, Raissadati A, Heinrich P, Wu SM. The cardiac conduction system: History, development, and disease. Curr Top Dev Biol 2024; 156:157-200. [PMID: 38556422 DOI: 10.1016/bs.ctdb.2024.02.006] [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] [Indexed: 04/02/2024]
Abstract
The heart is the first organ to form during embryonic development, establishing the circulatory infrastructure necessary to sustain life and enable downstream organogenesis. Critical to the heart's function is its ability to initiate and propagate electrical impulses that allow for the coordinated contraction and relaxation of its chambers, and thus, the movement of blood and nutrients. Several specialized structures within the heart, collectively known as the cardiac conduction system (CCS), are responsible for this phenomenon. In this review, we discuss the discovery and scientific history of the mammalian cardiac conduction system as well as the key genes and transcription factors implicated in the formation of its major structures. We also describe known human diseases related to CCS development and explore existing challenges in the clinical context.
Collapse
Affiliation(s)
- Carissa Lee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Sidra Xu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Tahmina Samad
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States; Division of Pediatric Cardiology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States; Department of Pediatrics, Stanford University, Stanford, CA, United States
| | - William R Goodyer
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States; Division of Pediatric Cardiology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Alireza Raissadati
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States; Division of Pediatric Cardiology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Paul Heinrich
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States; Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Cardiology, Klinikum Rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Sean M Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, United States; Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, United States.
| |
Collapse
|
5
|
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
|
6
|
Cao Y, Zheng M, Sewani MA, Wang J. The miR-17-92 cluster in cardiac health and disease. Birth Defects Res 2024; 116:e2273. [PMID: 37984445 PMCID: PMC11418803 DOI: 10.1002/bdr2.2273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 11/22/2023]
Abstract
MicroRNAs (miRs) are small noncoding RNAs that play important roles in both physiological and pathological processes through post-transcriptional regulation. The miR-17-92 cluster includes six individual members: miR-17, miR-18a, miR-19a, miR-19b-1, miR-20a, and miR-92a-1. The miR-17-92 cluster has been extensively studied and reported to broadly function in cancer biology, immunology, neurology, pulmonology, and cardiology. This review focuses on its roles in heart development and cardiac diseases. We briefly introduce the nature of the miR-17-92 cluster and its crucial roles in both normal development and the pathogenesis of various diseases. We summarize the recent progress in understanding the versatile roles of miR-17-92 during cardiac development, regeneration, and aging. Additionally, we highlight the indispensable roles of the miR-17-92 cluster in pathogenesis and therapeutic potential in cardiac birth defects and adult cardiac diseases.
Collapse
Affiliation(s)
- Yuhan Cao
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Mingjie Zheng
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Maham A Sewani
- Department of BioSciences, Wiess School of Natural Sciences, Rice University, Houston, TX 77030, USA
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, The University of Texas, Houston, TX 77030, USA
| |
Collapse
|
7
|
van der Maarel LE, Christoffels VM. Development of the Cardiac Conduction System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:185-200. [PMID: 38884712 DOI: 10.1007/978-3-031-44087-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The electrical impulses that coordinate the sequential, rhythmic contractions of the atria and ventricles are initiated and tightly regulated by the specialized tissues of the cardiac conduction system. In the mature heart, these impulses are generated by the pacemaker cardiomyocytes of the sinoatrial node, propagated through the atria to the atrioventricular node where they are delayed and then rapidly propagated to the atrioventricular bundle, right and left bundle branches, and finally, the peripheral ventricular conduction system. Each of these specialized components arise by complex patterning events during embryonic development. This chapter addresses the origins and transcriptional networks and signaling pathways that drive the development and maintain the function of the cardiac conduction system.
Collapse
Affiliation(s)
- Lieve E van der Maarel
- Department of Medical Biology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
8
|
Marcoux E, Sosnowski D, Ninni S, Mackasey M, Cadrin-Tourigny J, Roberts JD, Olesen MS, Fatkin D, Nattel S. Genetic Atrial Cardiomyopathies: Common Features, Specific Differences, and Broader Relevance to Understanding Atrial Cardiomyopathy. Circ Arrhythm Electrophysiol 2023; 16:675-698. [PMID: 38018478 DOI: 10.1161/circep.123.003750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Atrial cardiomyopathy is a condition that causes electrical and contractile dysfunction of the atria, often along with structural and functional changes. Atrial cardiomyopathy most commonly occurs in conjunction with ventricular dysfunction, in which case it is difficult to discern the atrial features that are secondary to ventricular dysfunction from those that arise as a result of primary atrial abnormalities. Isolated atrial cardiomyopathy (atrial-selective cardiomyopathy [ASCM], with minimal or no ventricular function disturbance) is relatively uncommon and has most frequently been reported in association with deleterious rare genetic variants. The genes involved can affect proteins responsible for various biological functions, not necessarily limited to the heart but also involving extracardiac tissues. Atrial enlargement and atrial fibrillation are common complications of ASCM and are often the predominant clinical features. Despite progress in identifying disease-causing rare variants, an overarching understanding and approach to the molecular pathogenesis, phenotypic spectrum, and treatment of genetic ASCM is still lacking. In this review, we aim to analyze the literature relevant to genetic ASCM to understand the key features of this rather rare condition, as well as to identify distinct characteristics of ASCM and its arrhythmic complications that are related to specific genotypes. We outline the insights that have been gained using basic research models of genetic ASCM in vitro and in vivo and correlate these with patient outcomes. Finally, we provide suggestions for the future investigation of patients with genetic ASCM and improvements to basic scientific models and systems. Overall, a better understanding of the genetic underpinnings of ASCM will not only provide a better understanding of this condition but also promises to clarify our appreciation of the more commonly occurring forms of atrial cardiomyopathy associated with ventricular dysfunction.
Collapse
Affiliation(s)
- Edouard Marcoux
- Research Center, Montreal Heart Institute, Université de Montréal. (E.M., D.S., S. Ninni, M.M., S. Nattel)
- Faculty of Pharmacy, Université de Montréal. (E.M.)
| | - Deanna Sosnowski
- Research Center, Montreal Heart Institute, Université de Montréal. (E.M., D.S., S. Ninni, M.M., S. Nattel)
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (D.S., M.M., S. Nattel)
| | - Sandro Ninni
- Research Center, Montreal Heart Institute, Université de Montréal. (E.M., D.S., S. Ninni, M.M., S. Nattel)
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, France (S. Ninni)
| | - Martin Mackasey
- Research Center, Montreal Heart Institute, Université de Montréal. (E.M., D.S., S. Ninni, M.M., S. Nattel)
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (D.S., M.M., S. Nattel)
| | - Julia Cadrin-Tourigny
- Cardiovascular Genetics Center, Montreal Heart Institute, Faculty of Medicine, Université de Montréal. (J.C.-T.)
| | - Jason D Roberts
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Canada (J.D.R.)
| | - Morten Salling Olesen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (M.S.O.)
| | - Diane Fatkin
- Victor Chang Cardiac Research Institute, Darlinghurst (D.F.)
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington (D.F.)
- Department of Cardiology, St Vincent's Hospital, Darlinghurst, NSW, Australia (D.F.)
| | - Stanley Nattel
- Research Center, Montreal Heart Institute, Université de Montréal. (E.M., D.S., S. Ninni, M.M., S. Nattel)
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal. (S. Nattel.)
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (D.S., M.M., S. Nattel)
- Institute of Pharmacology. West German Heart and Vascular Center, University Duisburg-Essen, Germany (S. Nattel)
- IHU LYRIC & Fondation Bordeaux Université de Bordeaux, France (S. Nattel)
| |
Collapse
|
9
|
Asjad E, Dobrzynski H. MicroRNAs: Midfielders of Cardiac Health, Disease and Treatment. Int J Mol Sci 2023; 24:16207. [PMID: 38003397 PMCID: PMC10671258 DOI: 10.3390/ijms242216207] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/05/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNA molecules that play a role in post-transcriptional gene regulation. It is generally accepted that their main mechanism of action is the negative regulation of gene expression, through binding to specific regions in messenger RNA (mRNA) and repressing protein translation. By interrupting protein synthesis, miRNAs can effectively turn genes off and influence many basic processes in the body, such as developmental and apoptotic behaviours of cells and cardiac organogenesis. Their importance is highlighted by inhibiting or overexpressing certain miRNAs, which will be discussed in the context of coronary artery disease, atrial fibrillation, bradycardia, and heart failure. Dysregulated levels of miRNAs in the body can exacerbate or alleviate existing disease, and their omnipresence in the body makes them reliable as quantifiable markers of disease. This review aims to provide a summary of miRNAs as biomarkers and their interactions with targets that affect cardiac health, and intersperse it with current therapeutic knowledge. It intends to succinctly inform on these topics and guide readers toward more comprehensive works if they wish to explore further through a wide-ranging citation list.
Collapse
Affiliation(s)
- Emman Asjad
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK;
| | - Halina Dobrzynski
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK;
- Department of Anatomy, Jagiellonian University Medical College, 31-034 Krakow, Poland
| |
Collapse
|
10
|
Chen X, Tang H, Lu K, Niu Z, Sheng W, Hwang HY, Pang PYK, Phillips JD, Khoynezhad A, Qu X, Li B, Han W. Gene modules and genes associated with postoperative atrial fibrillation: weighted gene co-expression network analysis and circRNA-miRNA-mRNA regulatory network analysis. J Thorac Dis 2023; 15:4949-4960. [PMID: 37868904 PMCID: PMC10586969 DOI: 10.21037/jtd-23-1179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 08/31/2023] [Indexed: 10/24/2023]
Abstract
Background Atrial fibrillation (AF) is the most common complication in patients undergoing cardiac surgery. However, the pathogenesis of postoperative AF (POAF) is elusive, and research related to this topic is sparse. Our study aimed to identify key gene modules and genes and to conduct a circular RNA (circRNA)-microRNA (miRNA)-messenger RNA (mRNA) regulatory network analysis of POAF on the basis of bioinformatic analysis. Methods The GSE143924 and GSE97455 data sets from the Gene Expression Omnibus (GEO) database were analyzed. Weighted gene co-expression network analysis (WGCNA) was used to identify the key gene modules and genes related to POAF. A circRNA-miRNA-mRNA regulatory network was also built according to differential expression analysis. Functional enrichment analysis was further performed according to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. Results WGCNA identified 2 key gene modules and 44 key genes that were significantly related to POAF. Functional enrichment analysis of these key genes implicated the following important biological processes (BPs): endosomal transport, protein kinase B signaling, and transcription regulation. The circRNA-miRNA-mRNA regulatory network suggested that KLF10 may take critical part in POAF. Moreover, 2 novel circRNAs, hsa_circRNA_001654 and hsa_circRNA_005899, and 2 miRNAs, hsa-miR-19b-3p and hsa-miR-30a-5p, which related with KLF10, were involved in the network. Conclusions Our study provides foundational expression profiles following POAF based on WGCNA. The circRNA-miRNA-mRNA network offers insights into the BPs and underlying mechanisms of POAF.
Collapse
Affiliation(s)
- Xiaomeng Chen
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Department of Cardiology, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Huaiguang Tang
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Department of Cardiology, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Kongmiao Lu
- Department of Pulmonary and Critical Care Medicine, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Zhaozhuo Niu
- Department of Cardiovascular Surgery, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Wei Sheng
- Department of Cardiovascular Surgery, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Ho Young Hwang
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Philip Y. K. Pang
- Department of Cardiothoracic Surgery, National Heart Centre Singapore, Singapore, Singapore
| | - Joseph D. Phillips
- Section of Thoracic Surgery, Dartmouth-Hitchcock Medical Center, 1 Medical Center Dr., Lebanon, NH, USA
| | - Ali Khoynezhad
- Department of Cardiovascular Surgery, MemorialCare Heart and Vascular Institute, Long Beach, CA, USA
| | - Xiaolu Qu
- Department of Pulmonary and Critical Care Medicine, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Bingong Li
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Department of Cardiology, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Wei Han
- Department of Pulmonary and Critical Care Medicine, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
11
|
Schulz C, Lemoine MD, Mearini G, Koivumäki J, Sani J, Schwedhelm E, Kirchhof P, Ghalawinji A, Stoll M, Hansen A, Eschenhagen T, Christ T. PITX2 Knockout Induces Key Findings of Electrical Remodeling as Seen in Persistent Atrial Fibrillation. Circ Arrhythm Electrophysiol 2023; 16:e011602. [PMID: 36763906 DOI: 10.1161/circep.122.011602] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
BACKGROUND Electrical remodeling in human persistent atrial fibrillation is believed to result from rapid electrical activation of the atria, but underlying genetic causes may contribute. Indeed, common gene variants in an enhancer region close to PITX2 (paired-like homeodomain transcription factor 2) are strongly associated with atrial fibrillation, but the mechanism behind this association remains unknown. This study evaluated the consequences of PITX2 deletion (PITX2-/-) in human induced pluripotent stem cell-derived atrial cardiomyocytes. METHODS CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9) was used to delete PITX2 in a healthy human iPSC line that served as isogenic control. Human induced pluripotent stem cell-derived atrial cardiomyocytes were differentiated with unfiltered retinoic acid and cultured in atrial engineered heart tissue. Force and action potential were measured in atrial engineered heart tissues. Single human induced pluripotent stem cell-derived atrial cardiomyocytes were isolated from atrial engineered heart tissue for ion current measurements. RESULTS PITX2-/- atrial engineered heart tissue beats slightly slower than isogenic control without irregularity. Force was lower in PITX2-/- than in isogenic control (0.053±0.015 versus 0.131±0.017 mN, n=28/3 versus n=28/4, PITX2-/- versus isogenic control; P<0.0001), accompanied by lower expression of CACNA1C and lower L-type Ca2+ current density. Early repolarization was weaker (action potential duration at 20% repolarization; 45.5±13.2 versus 8.6±5.3 ms, n=18/3 versus n=12/4, PITX2-/- versus isogenic control; P<0.0001), and maximum diastolic potential was more negative (-78.3±3.1 versus -69.7±0.6 mV, n=18/3 versus n=12/4, PITX2-/- versus isogenic control; P=0.001), despite normal inward rectifier currents (both IK1 and IK,ACh) and carbachol-induced shortening of action potential duration. CONCLUSIONS Complete PITX2 deficiency in human induced pluripotent stem cell-derived atrial cardiomyocytes recapitulates some findings of electrical remodeling of atrial fibrillation in the absence of fast beating, indicating that these abnormalities could be primary consequences of lower PITX2 levels.
Collapse
Affiliation(s)
- Carl Schulz
- Institute of Experimental Pharmacology and Toxicology (C.S., M.D.L., G.M., J.S., A.H., T.E., T.C.), University Medical Center Hamburg-Eppendorf, Germany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck (C.S., M.D.L., G.M., J.S., E.S., P.K.)
| | - Marc D Lemoine
- Institute of Experimental Pharmacology and Toxicology (C.S., M.D.L., G.M., J.S., A.H., T.E., T.C.), University Medical Center Hamburg-Eppendorf, Germany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck (C.S., M.D.L., G.M., J.S., E.S., P.K.)
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany (M.D.L., A.H., P.K., T.E., T.C.)
| | - Giulia Mearini
- Institute of Experimental Pharmacology and Toxicology (C.S., M.D.L., G.M., J.S., A.H., T.E., T.C.), University Medical Center Hamburg-Eppendorf, Germany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck (C.S., M.D.L., G.M., J.S., E.S., P.K.)
- DiNAQOR AG, Pfäffikon, Switzerland (G.M., P.K.)
| | - Jussi Koivumäki
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Finland (J.K.)
| | - Jascha Sani
- Institute of Experimental Pharmacology and Toxicology (C.S., M.D.L., G.M., J.S., A.H., T.E., T.C.), University Medical Center Hamburg-Eppendorf, Germany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck (C.S., M.D.L., G.M., J.S., E.S., P.K.)
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology (E.S.), University Medical Center Hamburg-Eppendorf, Germany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck (C.S., M.D.L., G.M., J.S., E.S., P.K.)
| | - Paulus Kirchhof
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck (C.S., M.D.L., G.M., J.S., E.S., P.K.)
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany (M.D.L., A.H., P.K., T.E., T.C.)
- DiNAQOR AG, Pfäffikon, Switzerland (G.M., P.K.)
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (P.K.)
| | - Amer Ghalawinji
- Division of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Germany (A.G., M.S.)
| | - Monika Stoll
- Division of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Germany (A.G., M.S.)
- Department of Biochemistry, CARIM School for Cardiovascular Sciences, Maastricht University, the Netherlands (M.S.)
| | - Arne Hansen
- Institute of Experimental Pharmacology and Toxicology (C.S., M.D.L., G.M., J.S., A.H., T.E., T.C.), University Medical Center Hamburg-Eppendorf, Germany
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany (M.D.L., A.H., P.K., T.E., T.C.)
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology (C.S., M.D.L., G.M., J.S., A.H., T.E., T.C.), University Medical Center Hamburg-Eppendorf, Germany
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany (M.D.L., A.H., P.K., T.E., T.C.)
| | - Torsten Christ
- Institute of Experimental Pharmacology and Toxicology (C.S., M.D.L., G.M., J.S., A.H., T.E., T.C.), University Medical Center Hamburg-Eppendorf, Germany
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany (M.D.L., A.H., P.K., T.E., T.C.)
| |
Collapse
|
12
|
Nopp S, van der Bent ML, Kraemmer D, Königsbrügge O, Wojta J, Pabinger I, Ay C, Nossent AY. Circulatory miR-411-5p as a Novel Prognostic Biomarker for Major Adverse Cardiovascular Events in Patients with Atrial Fibrillation. Int J Mol Sci 2023; 24:3861. [PMID: 36835272 PMCID: PMC9964230 DOI: 10.3390/ijms24043861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/12/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
The risk stratification of patients with atrial fibrillation (AF) for subsequent cardiovascular events could help in guiding prevention strategies. In this study, we aimed at investigating circulating microRNAs as prognostic biomarkers for major adverse cardiovascular events (MACE) in AF patients. We conducted a three-stage nested case-control study within the framework of a prospective registry, including 347 AF patients. First, total small RNA-sequencing was performed in 26 patients (13 cases with MACE) and the differential expression of microRNAs was analyzed. Seven candidate microRNAs with promising results in a subgroup analysis on cardiovascular death were selected and measured via using RT-qPCR in 97 patients (42 cases with cardiovascular death). To further validate our findings and investigate broader clinical applicability, we analyzed the same microRNAs in a subsequent nested case-control study of 102 patients (37 cases with early MACE) by using Cox regression. In the microRNA discovery cohort (n = 26), we detected 184 well-expressed microRNAs in circulation without overt differential expression between the cases and controls. A subgroup analysis on cardiovascular death revealed 26 microRNAs that were differentially expressed at a significance level < 0.05 (three of which with an FDR-adjusted p-value <0.05). We, therefore, proceeded with a nested case-control approach (n = 97) focusing on patients with cardiovascular death and selected, in total, seven microRNAs for further RT-qPCR analysis. One microRNA, miR-411-5p, was significantly associated with cardiovascular death (adjusted HR (95% CI): 1.95 (1.04-3.67)). Further validation (n = 102) in patients who developed early MACE showed similar results (adjusted HR (95% CI) 2.35 (1.17-4.73)). In conclusion, circulating miR-411-5p could be a valuable prognostic biomarker for MACE in AF patients.
Collapse
Affiliation(s)
- Stephan Nopp
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - M. Leontien van der Bent
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 Leiden, The Netherlands
| | - Daniel Kraemmer
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - Oliver Königsbrügge
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - Johann Wojta
- Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, 1090 Vienna, Austria
| | - Ingrid Pabinger
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - Cihan Ay
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - Anne Yaël Nossent
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 Leiden, The Netherlands
- Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
13
|
Zheng M, Li RG, Song J, Zhao X, Tang L, Erhardt S, Chen W, Nguyen BH, Li X, Li M, Wang J, Evans SM, Christoffels VM, Li N, Wang J. Hippo-Yap Signaling Maintains Sinoatrial Node Homeostasis. Circulation 2022; 146:1694-1711. [PMID: 36317529 PMCID: PMC9897204 DOI: 10.1161/circulationaha.121.058777] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 09/20/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND The sinoatrial node (SAN) functions as the pacemaker of the heart, initiating rhythmic heartbeats. Despite its importance, the SAN is one of the most poorly understood cardiac entities because of its small size and complex composition and function. The Hippo signaling pathway is a molecular signaling pathway fundamental to heart development and regeneration. Although abnormalities of the Hippo pathway are associated with cardiac arrhythmias in human patients, the role of this pathway in the SAN is unknown. METHODS We investigated key regulators of the Hippo pathway in SAN pacemaker cells by conditionally inactivating the Hippo signaling kinases Lats1 and Lats2 using the tamoxifen-inducible, cardiac conduction system-specific Cre driver Hcn4CreERT2 with Lats1 and Lats2 conditional knockout alleles. In addition, the Hippo-signaling effectors Yap and Taz were conditionally inactivated in the SAN. To determine the function of Hippo signaling in the SAN and other cardiac conduction system components, we conducted a series of physiological and molecular experiments, including telemetry ECG recording, echocardiography, Masson Trichrome staining, calcium imaging, immunostaining, RNAscope, cleavage under targets and tagmentation sequencing using antibodies against Yap1 or H3K4me3, quantitative real-time polymerase chain reaction, and Western blotting. We also performed comprehensive bioinformatics analyses of various datasets. RESULTS We found that Lats1/2 inactivation caused severe sinus node dysfunction. Compared with the controls, Lats1/2 conditional knockout mutants exhibited dysregulated calcium handling and increased fibrosis in the SAN, indicating that Lats1/2 function through both cell-autonomous and non-cell-autonomous mechanisms. It is notable that the Lats1/2 conditional knockout phenotype was rescued by genetic deletion of Yap and Taz in the cardiac conduction system. These rescued mice had normal sinus rhythm and reduced fibrosis of the SAN, indicating that Lats1/2 function through Yap and Taz. Cleavage Under Targets and Tagmentation sequencing data showed that Yap potentially regulates genes critical for calcium homeostasis such as Ryr2 and genes encoding paracrine factors important in intercellular communication and fibrosis induction such as Tgfb1 and Tgfb3. Consistent with this, Lats1/2 conditional knockout mutants had decreased Ryr2 expression and increased Tgfb1 and Tgfb3 expression compared with control mice. CONCLUSIONS We reveal, for the first time to our knowledge, that the canonical Hippo-Yap pathway plays a pivotal role in maintaining SAN homeostasis.
Collapse
Affiliation(s)
- Mingjie Zheng
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (M.Z., X.Z., S.E., W.C., Jun Wang)
| | - Rich G Li
- Texas Heart Institute, Houston (R.G.L., X.L.)
| | - Jia Song
- Department of Medicine (Section of Cardiovascular Research), Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (J.S., N.L.)
| | - Xiaolei Zhao
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (M.Z., X.Z., S.E., W.C., Jun Wang)
| | - Li Tang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan, China (L.T., M.L., Jianxin Wang)
| | - Shannon Erhardt
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (M.Z., X.Z., S.E., W.C., Jun Wang)
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, The University of Texas, Houston (S.E., Jun Wang)
| | - Wen Chen
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (M.Z., X.Z., S.E., W.C., Jun Wang)
| | - Bao H Nguyen
- Department of Molecular Physiology and Biophysics (B.H.N.)
| | - Xiao Li
- Texas Heart Institute, Houston (R.G.L., X.L.)
| | - Min Li
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan, China (L.T., M.L., Jianxin Wang)
| | - Jianxin Wang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan, China (L.T., M.L., Jianxin Wang)
| | - Sylvia M Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Departments of Pharmacology and Medicine, University of California at San Diego, La Jolla (S.M.E.)
| | - Vincent M Christoffels
- Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, The Netherlands (V.M.C.)
| | - Na Li
- Department of Medicine (Section of Cardiovascular Research), Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (J.S., N.L.)
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (M.Z., X.Z., S.E., W.C., Jun Wang)
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, The University of Texas, Houston (S.E., Jun Wang)
| |
Collapse
|
14
|
Su KN, Ma Y, Cacheux M, Ilkan Z, Raad N, Muller GK, Wu X, Guerrera N, Thorn SL, Sinusas AJ, Foretz M, Viollet B, Akar JG, Akar FG, Young LH. Atrial AMP-activated protein kinase is critical for prevention of dysregulation of electrical excitability and atrial fibrillation. JCI Insight 2022; 7:141213. [PMID: 35451373 PMCID: PMC9089788 DOI: 10.1172/jci.insight.141213] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 02/23/2022] [Indexed: 12/03/2022] Open
Abstract
Metabolic stress is an important cause of pathological atrial remodeling and atrial fibrillation. AMPK is a ubiquitous master metabolic regulator, yet its biological function in the atria is poorly understood in both health and disease. We investigated the impact of atrium-selective cardiac AMPK deletion on electrophysiological and structural remodeling in mice. Loss of atrial AMPK expression caused atrial changes in electrophysiological properties and atrial ectopic activity prior to the onset of spontaneous atrial fibrillation. Concomitant transcriptional downregulation of connexins and atrial ion channel subunits manifested with delayed left atrial activation and repolarization. The early molecular and electrophysiological abnormalities preceded left atrial structural remodeling and interstitial fibrosis. AMPK inactivation induced downregulation of transcription factors (Mef2c and Pitx2c) linked to connexin and ion channel transcriptional reprogramming. Thus, AMPK plays an essential homeostatic role in atria, protecting against adverse remodeling potentially by regulating key transcription factors that control the expression of atrial ion channels and gap junction proteins.
Collapse
Affiliation(s)
- Kevin N Su
- Department of Cellular & Molecular Physiology and
| | - Yina Ma
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marine Cacheux
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Zeki Ilkan
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nour Raad
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Xiaohong Wu
- Department of Cellular & Molecular Physiology and
| | - Nicole Guerrera
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Stephanie L Thorn
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Albert J Sinusas
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marc Foretz
- Institut Cochin, Université de Paris, CNRS, INSERM, Paris, France
| | - Benoit Viollet
- Institut Cochin, Université de Paris, CNRS, INSERM, Paris, France
| | - Joseph G Akar
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Fadi G Akar
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lawrence H Young
- Department of Cellular & Molecular Physiology and.,Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
15
|
Li H, Zhan J, Chen C, Wang D. MicroRNAs in cardiovascular diseases. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:140-168. [PMID: 37724243 PMCID: PMC10471109 DOI: 10.1515/mr-2021-0001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 12/29/2021] [Indexed: 09/20/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading causes of death and disability worldwide, despite the wide diversity of molecular targets identified and the development of therapeutic methods. MicroRNAs (miRNAs) are a class of small (about 22 nucleotides) non-coding RNAs (ncRNAs) that negatively regulate gene expression at the post-transcriptional level in the cytoplasm and play complicated roles in different CVDs. While miRNA overexpression in one type of cell protects against heart disease, it promotes cardiac dysfunction in another type of cardiac cell. Moreover, recent studies have shown that, apart from cytosolic miRNAs, subcellular miRNAs such as mitochondria- and nucleus-localized miRNAs are dysregulated in CVDs. However, the functional properties of cellular- and subcellular-localized miRNAs have not been well characterized. In this review article, by carefully revisiting animal-based miRNA studies in CVDs, we will address the regulation and functional properties of miRNAs in various CVDs. Specifically, the cell-cell crosstalk and subcellular perspective of miRNAs are highlighted. We will provide the background for attractive molecular targets that might be useful in preventing the progression of CVDs and heart failure (HF) as well as insights for future studies.
Collapse
Affiliation(s)
- Huaping Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Jiabing Zhan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Daowen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| |
Collapse
|
16
|
Lozano-Velasco E, Garcia-Padilla C, del Mar Muñoz-Gallardo M, Martinez-Amaro FJ, Caño-Carrillo S, Castillo-Casas JM, Sanchez-Fernandez C, Aranega AE, Franco D. Post-Transcriptional Regulation of Molecular Determinants during Cardiogenesis. Int J Mol Sci 2022; 23:ijms23052839. [PMID: 35269981 PMCID: PMC8911333 DOI: 10.3390/ijms23052839] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/19/2022] [Accepted: 02/26/2022] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular development is initiated soon after gastrulation as bilateral precardiac mesoderm is progressively symmetrically determined at both sides of the developing embryo. The precardiac mesoderm subsequently fused at the embryonic midline constituting an embryonic linear heart tube. As development progress, the embryonic heart displays the first sign of left-right asymmetric morphology by the invariably rightward looping of the initial heart tube and prospective embryonic ventricular and atrial chambers emerged. As cardiac development progresses, the atrial and ventricular chambers enlarged and distinct left and right compartments emerge as consequence of the formation of the interatrial and interventricular septa, respectively. The last steps of cardiac morphogenesis are represented by the completion of atrial and ventricular septation, resulting in the configuration of a double circuitry with distinct systemic and pulmonary chambers, each of them with distinct inlets and outlets connections. Over the last decade, our understanding of the contribution of multiple growth factor signaling cascades such as Tgf-beta, Bmp and Wnt signaling as well as of transcriptional regulators to cardiac morphogenesis have greatly enlarged. Recently, a novel layer of complexity has emerged with the discovery of non-coding RNAs, particularly microRNAs and lncRNAs. Herein, we provide a state-of-the-art review of the contribution of non-coding RNAs during cardiac development. microRNAs and lncRNAs have been reported to functional modulate all stages of cardiac morphogenesis, spanning from lateral plate mesoderm formation to outflow tract septation, by modulating major growth factor signaling pathways as well as those transcriptional regulators involved in cardiac development.
Collapse
Affiliation(s)
- Estefania Lozano-Velasco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
- Fundación Medina, 18007 Granada, Spain
| | - Carlos Garcia-Padilla
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
- Department of Anatomy, Embryology and Zoology, School of Medicine, University of Extremadura, 06006 Badajoz, Spain
| | - Maria del Mar Muñoz-Gallardo
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
| | - Francisco Jose Martinez-Amaro
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
| | - Sheila Caño-Carrillo
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
| | - Juan Manuel Castillo-Casas
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
| | - Cristina Sanchez-Fernandez
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
- Fundación Medina, 18007 Granada, Spain
| | - Amelia E. Aranega
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
- Fundación Medina, 18007 Granada, Spain
| | - Diego Franco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
- Fundación Medina, 18007 Granada, Spain
- Correspondence:
| |
Collapse
|
17
|
van Ouwerkerk AF, Bosada FM, van Duijvenboden K, Houweling AC, Scholman KT, Wakker V, Allaart CP, Uhm JS, Mathijssen IB, Baartscheer T, Postma AV, Barnett P, Verkerk AO, Boukens BJ, Christoffels VM. Patient-specific TBX5-G125R Variant Induces Profound Transcriptional Deregulation and Atrial Dysfunction. Circulation 2022; 145:606-619. [PMID: 35113653 PMCID: PMC8860223 DOI: 10.1161/circulationaha.121.054347] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: The pathogenic missense variant p.G125R in TBX5 causes Holt-Oram syndrome (HOS; hand-heart syndrome) and early onset of atrial fibrillation. Revealing how an altered key developmental transcription factor modulates cardiac physiology in vivo will provide unique insights into the mechanisms underlying atrial fibrillation in these patients. Methods: We analyzed electrocardiograms (ECGs) of an extended family pedigree of HOS patients. Next, we introduced the TBX5-p.G125R variant in the mouse genome (Tbx5G125R) and performed electrophysiological analyses (ECG, optical mapping, patch clamp, intracellular calcium measurements), transcriptomics (single nuclei and tissue RNA sequencing) and epigenetic profiling (ATAC-sequencing, H3K27ac CUT&RUN-sequencing). Results: We discovered high incidence of atrial extra systoles and atrioventricular conduction disturbances in HOS patients. Tbx5G125R/+ mice were morphologically unaffected and displayed variable RR intervals, atrial extra systoles and susceptibility to atrial fibrillation, reminiscent of TBX5-p.G125R patients. Atrial conduction velocity was not affected but systolic and diastolic intracellular calcium concentrations were decreased and action potentials prolonged in isolated cardiomyocytes of Tbx5G125R/+ mice compared to controls. Transcriptional profiling of atria revealed most profound transcriptional changes in cardiomyocytes versus other cell types, and identified over a thousand coding and non-coding transcripts that were differentially expressed. Epigenetic profiling uncovered thousands of TBX5-p.G125R sensitive putative regulatory elements (including enhancers) that gained accessibility in atrial cardiomyocytes. The majority of sites with increased accessibility were occupied by Tbx5. The small group of sites with reduced accessibility was enriched for DNA binding motifs of members of the SP- and KLF families of transcription factors. These data show that Tbx5-p.G125R induces changes in regulatory element activity, altered transcriptional regulation and changed cardiomyocyte behavior, possibly caused by altered DNA binding and cooperativity properties. Conclusions: Our data reveal how a disease-causing missense variant in TBX5 induces profound changes in the atrial transcriptional regulatory network and epigenetic state in vivo, leading to arrhythmia reminiscent of those seen in human TBX5-p.G125R variant carriers.
Collapse
Affiliation(s)
- Antoinette F van Ouwerkerk
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands; Aix-Marseille University, INSERM, TAGC, U1090, Marseille, France
| | - Fernanda M Bosada
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Karel van Duijvenboden
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Arjan C Houweling
- Department of Human Genetics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Koen T Scholman
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Vincent Wakker
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Cornelis P Allaart
- Department of Cardiology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Jae-Sun Uhm
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Inge B Mathijssen
- Department of Human Genetics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Ton Baartscheer
- Department of Experimental Cardiology, Amsterdam University Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Alex V Postma
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands; Department of Human Genetics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Phil Barnett
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands; Department of Experimental Cardiology, Amsterdam University Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Bastiaan J Boukens
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Li D, Nie J, Han Y, Ni L. Epigenetic Mechanism and Therapeutic Implications of Atrial Fibrillation. Front Cardiovasc Med 2022; 8:763824. [PMID: 35127848 PMCID: PMC8815458 DOI: 10.3389/fcvm.2021.763824] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/08/2021] [Indexed: 12/28/2022] Open
Abstract
Atrial fibrillation (AF) is the most common arrhythmia attacking 1. 5–2.0% of general population worldwide. It has a significant impact on morbidity and mortality globally and its prevalence increases exponentially with age. Therapies like catheter ablation or conventional antiarrhythmic drugs have not provided effective solution to the recurrence for AF over the past decades. Over 100 genetic loci have been discovered to be associated with AF by Genome-wide association studies (GWAS) but none has led to a therapy. Recently potential involvement of epigenetics (DNA methylation, histone modification, and non-coding RNAs) in the initiation and maintenance of AF has partly emerged as proof-of-concept in the mechanism and management of AF. Here we reviewed the epigenetic features involved in AF pathophysiology and provided an update of their implications in AF therapy.
Collapse
|
19
|
Hu S, Mahadevan A, Elysee IF, Choi J, Souchet NR, Bae GH, Taboada AK, Sanketi B, Duhamel GE, Sevier CS, Tao G, Kurpios NA. The asymmetric Pitx2 gene regulates gut muscular-lacteal development and protects against fatty liver disease. Cell Rep 2021; 37:110030. [PMID: 34818545 PMCID: PMC8650168 DOI: 10.1016/j.celrep.2021.110030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 08/19/2021] [Accepted: 10/29/2021] [Indexed: 12/25/2022] Open
Abstract
Intestinal lacteals are essential lymphatic channels for absorption and transport of dietary lipids and drive the pathogenesis of debilitating metabolic diseases. However, organ-specific mechanisms linking lymphatic dysfunction to disease etiology remain largely unknown. In this study, we uncover an intestinal lymphatic program that is linked to the left-right (LR) asymmetric transcription factor Pitx2. We show that deletion of the asymmetric Pitx2 enhancer ASE alters normal lacteal development through the lacteal-associated contractile smooth muscle lineage. ASE deletion leads to abnormal muscle morphogenesis induced by oxidative stress, resulting in impaired lacteal extension and defective lymphatic system-dependent lipid transport. Surprisingly, activation of lymphatic system-independent trafficking directs dietary lipids from the gut directly to the liver, causing diet-induced fatty liver disease. Our study reveals the molecular mechanism linking gut lymphatic function to the earliest symmetry-breaking Pitx2 and highlights the important relationship between intestinal lymphangiogenesis and the gut-liver axis.
Collapse
Affiliation(s)
- Shing Hu
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Aparna Mahadevan
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Isaac F Elysee
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Joseph Choi
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Nathan R Souchet
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Gloria H Bae
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Alessandra K Taboada
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Bhargav Sanketi
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Gerald E Duhamel
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Carolyn S Sevier
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Ge Tao
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA.
| |
Collapse
|
20
|
Iop L, Iliceto S, Civieri G, Tona F. Inherited and Acquired Rhythm Disturbances in Sick Sinus Syndrome, Brugada Syndrome, and Atrial Fibrillation: Lessons from Preclinical Modeling. Cells 2021; 10:3175. [PMID: 34831398 PMCID: PMC8623957 DOI: 10.3390/cells10113175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Rhythm disturbances are life-threatening cardiovascular diseases, accounting for many deaths annually worldwide. Abnormal electrical activity might arise in a structurally normal heart in response to specific triggers or as a consequence of cardiac tissue alterations, in both cases with catastrophic consequences on heart global functioning. Preclinical modeling by recapitulating human pathophysiology of rhythm disturbances is fundamental to increase the comprehension of these diseases and propose effective strategies for their prevention, diagnosis, and clinical management. In silico, in vivo, and in vitro models found variable application to dissect many congenital and acquired rhythm disturbances. In the copious list of rhythm disturbances, diseases of the conduction system, as sick sinus syndrome, Brugada syndrome, and atrial fibrillation, have found extensive preclinical modeling. In addition, the electrical remodeling as a result of other cardiovascular diseases has also been investigated in models of hypertrophic cardiomyopathy, cardiac fibrosis, as well as arrhythmias induced by other non-cardiac pathologies, stress, and drug cardiotoxicity. This review aims to offer a critical overview on the effective ability of in silico bioinformatic tools, in vivo animal studies, in vitro models to provide insights on human heart rhythm pathophysiology in case of sick sinus syndrome, Brugada syndrome, and atrial fibrillation and advance their safe and successful translation into the cardiology arena.
Collapse
Affiliation(s)
- Laura Iop
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padua, Via Giustiniani, 2, I-35124 Padua, Italy; (S.I.); (G.C.)
| | | | | | - Francesco Tona
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padua, Via Giustiniani, 2, I-35124 Padua, Italy; (S.I.); (G.C.)
| |
Collapse
|
21
|
Zhang L, Wang X, Huang C. A narrative review of non-coding RNAs in atrial fibrillation: potential therapeutic targets and molecular mechanisms. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1486. [PMID: 34734038 PMCID: PMC8506732 DOI: 10.21037/atm-21-4483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/16/2021] [Indexed: 11/11/2022]
Abstract
Objective This review summarizes the advances in the study of ncRNAs and atrial remodeling mechanisms to explore potential therapeutic targets and strategies for AF. Background Atrial fibrillation (AF) is one of the most common arrhythmias, and its morbidity and mortality rates are gradually increasing. Non-coding ribonucleic acid RNAs (ncRNAs) are transcribed from the genome and do not have the ability to be translated into proteins. A growing body of evidence has shown ncRNAs are extensively involved in the pathophysiological processes underlying AF. However, the precise molecular mechanisms of these associations have not been fully elucidated. Atrial remodeling plays a key role in the occurrence and development of AF, and includes electrical remodeling, structural remodeling, and autonomic nerve remodeling. Research has shown that ncRNA expression is altered in the plasma and tissues of AF patients that mediate cardiac excitation and arrhythmia, and is closely related to atrial remodeling. Methods Literatures about ncRNAs and atrial fibrillation were extensively reviewed to discuss and analyze. Conclusions The biology of ncRNAs represents a relatively new field of research and is still in an emerging stage. Recent studies have laid a foundation for understanding the molecular mechanisms of AF, future studies aimed at identifying how ncRNAs act on atrial fibrillation to provide potentially promising therapeutic targets for the treatment of atrial fibrillation.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
22
|
A microRNA program regulates the balance between cardiomyocyte hyperplasia and hypertrophy and stimulates cardiac regeneration. Nat Commun 2021; 12:4808. [PMID: 34376683 PMCID: PMC8355162 DOI: 10.1038/s41467-021-25211-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 07/28/2021] [Indexed: 11/09/2022] Open
Abstract
Myocardial regeneration is restricted to early postnatal life, when mammalian cardiomyocytes still retain the ability to proliferate. The molecular cues that induce cell cycle arrest of neonatal cardiomyocytes towards terminally differentiated adult heart muscle cells remain obscure. Here we report that the miR-106b~25 cluster is higher expressed in the early postnatal myocardium and decreases in expression towards adulthood, especially under conditions of overload, and orchestrates the transition of cardiomyocyte hyperplasia towards cell cycle arrest and hypertrophy by virtue of its targetome. In line, gene delivery of miR-106b~25 to the mouse heart provokes cardiomyocyte proliferation by targeting a network of negative cell cycle regulators including E2f5, Cdkn1c, Ccne1 and Wee1. Conversely, gene-targeted miR-106b~25 null mice display spontaneous hypertrophic remodeling and exaggerated remodeling to overload by derepression of the prohypertrophic transcription factors Hand2 and Mef2d. Taking advantage of the regulatory function of miR-106b~25 on cardiomyocyte hyperplasia and hypertrophy, viral gene delivery of miR-106b~25 provokes nearly complete regeneration of the adult myocardium after ischemic injury. Our data demonstrate that exploitation of conserved molecular programs can enhance the regenerative capacity of the injured heart.
Collapse
|
23
|
Bai J, Lu Y, Zhu Y, Wang H, Yin D, Zhang H, Franco D, Zhao J. Understanding PITX2-Dependent Atrial Fibrillation Mechanisms through Computational Models. Int J Mol Sci 2021; 22:7681. [PMID: 34299303 PMCID: PMC8307824 DOI: 10.3390/ijms22147681] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 01/11/2023] Open
Abstract
Atrial fibrillation (AF) is a common arrhythmia. Better prevention and treatment of AF are needed to reduce AF-associated morbidity and mortality. Several major mechanisms cause AF in patients, including genetic predispositions to AF development. Genome-wide association studies have identified a number of genetic variants in association with AF populations, with the strongest hits clustering on chromosome 4q25, close to the gene for the homeobox transcription PITX2. Because of the inherent complexity of the human heart, experimental and basic research is insufficient for understanding the functional impacts of PITX2 variants on AF. Linking PITX2 properties to ion channels, cells, tissues, atriums and the whole heart, computational models provide a supplementary tool for achieving a quantitative understanding of the functional role of PITX2 in remodelling atrial structure and function to predispose to AF. It is hoped that computational approaches incorporating all we know about PITX2-related structural and electrical remodelling would provide better understanding into its proarrhythmic effects leading to development of improved anti-AF therapies. In the present review, we discuss advances in atrial modelling and focus on the mechanistic links between PITX2 and AF. Challenges in applying models for improving patient health are described, as well as a summary of future perspectives.
Collapse
Affiliation(s)
- Jieyun Bai
- College of Information Science and Technology, Jinan University, Guangzhou 510632, China; (Y.L.); (Y.Z.)
- Auckland Bioengineering Institute, University of Auckland, Auckland 1010, New Zealand
| | - Yaosheng Lu
- College of Information Science and Technology, Jinan University, Guangzhou 510632, China; (Y.L.); (Y.Z.)
| | - Yijie Zhu
- College of Information Science and Technology, Jinan University, Guangzhou 510632, China; (Y.L.); (Y.Z.)
| | - Huijin Wang
- College of Information Science and Technology, Jinan University, Guangzhou 510632, China; (Y.L.); (Y.Z.)
| | - Dechun Yin
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin 150000, China;
| | - Henggui Zhang
- Biological Physics Group, School of Physics & Astronomy, The University of Manchester, Manchester M13 9PL, UK;
| | - Diego Franco
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain;
| | - Jichao Zhao
- Auckland Bioengineering Institute, University of Auckland, Auckland 1010, New Zealand
| |
Collapse
|
24
|
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
|
25
|
Differential Spatio-Temporal Regulation of T-Box Gene Expression by microRNAs during Cardiac Development. J Cardiovasc Dev Dis 2021; 8:jcdd8050056. [PMID: 34068962 PMCID: PMC8156480 DOI: 10.3390/jcdd8050056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 01/05/2023] Open
Abstract
Cardiovascular development is a complex process that starts with the formation of symmetrically located precardiac mesodermal precursors soon after gastrulation and is completed with the formation of a four-chambered heart with distinct inlet and outlet connections. Multiple transcriptional inputs are required to provide adequate regional identity to the forming atrial and ventricular chambers as well as their flanking regions; i.e., inflow tract, atrioventricular canal, and outflow tract. In this context, regional chamber identity is widely governed by regional activation of distinct T-box family members. Over the last decade, novel layers of gene regulatory mechanisms have been discovered with the identification of non-coding RNAs. microRNAs represent the most well-studied subcategory among short non-coding RNAs. In this study, we sought to investigate the functional role of distinct microRNAs that are predicted to target T-box family members. Our data demonstrated a highly dynamic expression of distinct microRNAs and T-box family members during cardiogenesis, revealing a relatively large subset of complementary and similar microRNA-mRNA expression profiles. Over-expression analyses demonstrated that a given microRNA can distinctly regulate the same T-box family member in distinct cardiac regions and within distinct temporal frameworks, supporting the notion of indirect regulatory mechanisms, and dual luciferase assays on Tbx2, Tbx3 and Tbx5 3' UTR further supported this notion. Overall, our data demonstrated a highly dynamic microRNA and T-box family members expression during cardiogenesis and supported the notion that such microRNAs indirectly regulate the T-box family members in a tissue- and time-dependent manner.
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW Atrial fibrillation is the most common sustained cardiac arrhythmia. In addition to traditional risk factors, it is increasingly recognized that a genetic component underlies atrial fibrillation development. This review aims to provide an overview of the genetic cause of atrial fibrillation and clinical applications, with a focus on recent developments. RECENT FINDINGS Genome-wide association studies have now identified around 140 genetic loci associated with atrial fibrillation. Studies into the effects of several loci and their tentative gene targets have identified novel pathways associated with atrial fibrillation development. However, further validations of causality are still needed for many implicated genes. Genetic variants at identified loci also help predict individual atrial fibrillation risk and response to different therapies. SUMMARY Continued advances in the field of genetics and molecular biology have led to significant insight into the genetic underpinnings of atrial fibrillation. Potential clinical applications of these studies include the identification of new therapeutic targets and development of genetic risk scores to optimize management of this common cardiac arrhythmia.
Collapse
Affiliation(s)
- Jitae A. Kim
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mihail G. Chelu
- Department of Cardiology, Baylor College of Medicine, Houston, TX, USA
| | - Na Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX
| |
Collapse
|
27
|
Nattel S, Aguilar M. Do Atrial Fibrillation-Promoting Gene Variants Act by Enhancing Atrial Remodeling? JACC Clin Electrophysiol 2021; 6:1522-1524. [PMID: 33213812 DOI: 10.1016/j.jacep.2020.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Stanley Nattel
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Duisburg, Germany; Institut Hospitalo Universitaire de rythmologie et de modélisation cardiaque and Fondation Bordeaux Université, Bordeaux, France.
| | - Martin Aguilar
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
28
|
Disease Modeling and Disease Gene Discovery in Cardiomyopathies: A Molecular Study of Induced Pluripotent Stem Cell Generated Cardiomyocytes. Int J Mol Sci 2021; 22:ijms22073311. [PMID: 33805011 PMCID: PMC8037452 DOI: 10.3390/ijms22073311] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 01/04/2023] Open
Abstract
The in vitro modeling of cardiac development and cardiomyopathies in human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMs) provides opportunities to aid the discovery of genetic, molecular, and developmental changes that are causal to, or influence, cardiomyopathies and related diseases. To better understand the functional and disease modeling potential of iPSC-differentiated CMs and to provide a proof of principle for large, epidemiological-scale disease gene discovery approaches into cardiomyopathies, well-characterized CMs, generated from validated iPSCs of 12 individuals who belong to four sibships, and one of whom reported a major adverse cardiac event (MACE), were analyzed by genome-wide mRNA sequencing. The generated CMs expressed CM-specific genes and were highly concordant in their total expressed transcriptome across the 12 samples (correlation coefficient at 95% CI =0.92 ± 0.02). The functional annotation and enrichment analysis of the 2116 genes that were significantly upregulated in CMs suggest that generated CMs have a transcriptomic and functional profile of immature atrial-like CMs; however, the CMs-upregulated transcriptome also showed high overlap and significant enrichment in primary cardiomyocyte (p-value = 4.36 × 10−9), primary heart tissue (p-value = 1.37 × 10−41) and cardiomyopathy (p-value = 1.13 × 10−21) associated gene sets. Modeling the effect of MACE in the generated CMs-upregulated transcriptome identified gene expression phenotypes consistent with the predisposition of the MACE-affected sibship to arrhythmia, prothrombotic, and atherosclerosis risk.
Collapse
|
29
|
Abstract
Cardiac development is a complex developmental process that is initiated soon after gastrulation, as two sets of precardiac mesodermal precursors are symmetrically located and subsequently fused at the embryonic midline forming the cardiac straight tube. Thereafter, the cardiac straight tube invariably bends to the right, configuring the first sign of morphological left–right asymmetry and soon thereafter the atrial and ventricular chambers are formed, expanded and progressively septated. As a consequence of all these morphogenetic processes, the fetal heart acquired a four-chambered structure having distinct inlet and outlet connections and a specialized conduction system capable of directing the electrical impulse within the fully formed heart. Over the last decades, our understanding of the morphogenetic, cellular, and molecular pathways involved in cardiac development has exponentially grown. Multiples aspects of the initial discoveries during heart formation has served as guiding tools to understand the etiology of cardiac congenital anomalies and adult cardiac pathology, as well as to enlighten novels approaches to heal the damaged heart. In this review we provide an overview of the complex cellular and molecular pathways driving heart morphogenesis and how those discoveries have provided new roads into the genetic, clinical and therapeutic management of the diseased hearts.
Collapse
|
30
|
Zhang C, Chen X, Chen Y, Cao M, Tang J, Zhong B, He M. The PITX gene family as potential biomarkers and therapeutic targets in lung adenocarcinoma. Medicine (Baltimore) 2021; 100:e23936. [PMID: 33530195 PMCID: PMC7850728 DOI: 10.1097/md.0000000000023936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 07/06/2020] [Accepted: 11/25/2020] [Indexed: 01/05/2023] Open
Abstract
ABSTRACT The PITX gene family of transcription factors have been reported to regulate the development of multiple organs. This study was designed to investigate the role of PITXs in lung adenocarcinoma (LUAD).In this study, the transcriptional levels of the 3 identified PITXs in patients with LUAD were examined using the gene expression profiling interactive analysis interactive web server. Meanwhile, the immunohistochemical data of the 3 PITXs were obtained in the Human Protein Atlas website, and western blotting was additionally conducted for further verification. Moreover, the association between the levels of PITXs and the stage plot as well as overall survival of patients with LUAD was analyzed.We found that the mRNA and protein levels of PITX1 and PITX2 were higher in LUAD tissues than those in normal lung tissues, while those of PITX3 displayed no significant differences. Additionally, PITX1 and PITX3 were found to be significantly associated with the stage of LUAD. The Kaplan-Meier Plot showed that the high level of PITX1 conferred a better overall survival of patients with LUAD while the high level of PITX3 was associated with poor prognosis.Our study implied that PITX1 and PITX3 are potential targets of precision therapy for patients with LUAD while PITX1 and PITX2 are regarded as novel biomarkers for the diagnosis of LUAD.
Collapse
|
31
|
Oltra E. Epigenetics of muscle disorders. MEDICAL EPIGENETICS 2021:279-308. [DOI: 10.1016/b978-0-12-823928-5.00023-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
32
|
Bhattacharyya S, Munshi NV. Development of the Cardiac Conduction System. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037408. [PMID: 31988140 DOI: 10.1101/cshperspect.a037408] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The cardiac conduction system initiates and propagates each heartbeat. Specialized conducting cells are a well-conserved phenomenon across vertebrate evolution, although mammalian and avian species harbor specific components unique to organisms with four-chamber hearts. Early histological studies in mammals provided evidence for a dominant pacemaker within the right atrium and clarified the existence of the specialized muscular axis responsible for atrioventricular conduction. Building on these seminal observations, contemporary genetic techniques in a multitude of model organisms has characterized the developmental ontogeny, gene regulatory networks, and functional importance of individual anatomical compartments within the cardiac conduction system. This review describes in detail the transcriptional and regulatory networks that act during cardiac conduction system development and homeostasis with a particular emphasis on networks implicated in human electrical variation by large genome-wide association studies. We conclude with a discussion of the clinical implications of these studies and describe some future directions.
Collapse
Affiliation(s)
| | - Nikhil V Munshi
- Department of Internal Medicine, Division of Cardiology.,McDermott Center for Human Growth and Development.,Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas 75390, USA.,Hamon Center for Regenerative Science and Medicine, Dallas, Texas 75390, USA
| |
Collapse
|
33
|
Sohail H, Hassan SM, Yaqoob U, Hassan Z. The height as an independent risk factor of atrial fibrillation: A review. Indian Heart J 2020; 73:22-25. [PMID: 33714405 PMCID: PMC7961249 DOI: 10.1016/j.ihj.2020.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/16/2020] [Accepted: 11/07/2020] [Indexed: 12/04/2022] Open
Abstract
Atrial fibrillation (AF) is characterized by abnormal heart rhythm. Among other well-known associations, recent studies suggest an association of AF with height. Height is related to 50 diseases spanning different body systems, AF is one of them. Since AF, a heterogeneous disease process, is influenced by structural, neural, electrical, and hemodynamic factors, height alters this process through its contribution to increasing atrial and ventricular size, leading to altered conduction patterns, autonomic dysregulation, and development of AF. Multiple underlying mechanisms associate height with AF. Apart from these indirect mechanisms, genome-wide association studies suggest the involvement of the same genes in AF and growth pathways. Tall stature is independently associated with a higher risk of AF development in healthy individuals. Since adult height is achieved much earlier than the onset of AF, protective measures can be taken in individuals with increased height to monitor, manage, and prevent the progression of AF.
Collapse
Affiliation(s)
- Hamza Sohail
- Jinnah Sindh Medical University, Karachi, Pakistan.
| | | | - Uzair Yaqoob
- Dow University of Health Sciences, Karachi, Pakistan.
| | | |
Collapse
|
34
|
Zuccotti P, Peroni D, Potrich V, Quattrone A, Dassi E. Hyperconserved Elements in Human 5'UTRs Shape Essential Post-transcriptional Regulatory Networks. Front Mol Biosci 2020; 7:220. [PMID: 33005630 PMCID: PMC7484617 DOI: 10.3389/fmolb.2020.00220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 08/05/2020] [Indexed: 01/03/2023] Open
Abstract
Post-transcriptional regulation (PTR) of gene expression is a powerful determinant of cellular phenotypes. The 5′ and 3′ untranslated regions of the mRNA (UTRs) mediate this role through sequence and secondary structure elements bound by RNA-binding proteins (RBPs) and non-coding RNAs. While functional regions in the 3′UTRs have been extensively studied, the 5′UTRs are still relatively uncharacterized. To fill this gap, we used a computational approach exploiting phylogenetic conservation to identify hyperconserved elements in human 5′UTRs (5′HCEs). Our assumption was that 5′HCEs would represent evolutionarily stable and hence important PTR sites. We identified over 5000 5′HCEs occurring in 10% of human protein-coding genes. These sequence elements are rather short and mostly found in narrowly-spaced clusters. 5′HCEs-containing genes are enriched in essential cellular functions and include 20% of all homeotic genes. Homeotic genes are essential transcriptional regulators, driving body plan and neuromuscular development. However, the role of PTR in their expression is mostly unknown. By integrating computational and experimental approaches we identified RBMX as the initiator RBP of a post-transcriptional cascade regulating many homeotic genes. This work thus establishes 5′HCEs as mediators of essential post-transcriptional regulatory networks.
Collapse
Affiliation(s)
- Paola Zuccotti
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Daniele Peroni
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Valentina Potrich
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Alessandro Quattrone
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Erik Dassi
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| |
Collapse
|
35
|
Vaze A, Tran KV, Tanriverdi K, Sardana M, Lessard D, Donahue JK, Barton B, Aurigemma G, Lubitz SA, Lin H, Nasr GH, Mandapati A, Benjamin EJ, Vasan RS, Freedman JE, McManus DD. Relations between plasma microRNAs, echocardiographic markers of atrial remodeling, and atrial fibrillation: Data from the Framingham Offspring study. PLoS One 2020; 15:e0236960. [PMID: 32813736 PMCID: PMC7437902 DOI: 10.1371/journal.pone.0236960] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 07/16/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Circulating microRNAs may reflect or influence pathological cardiac remodeling and contribute to atrial fibrillation (AF). OBJECTIVE The purpose of this study was to identify candidate plasma microRNAs that are associated with echocardiographic phenotypes of atrial remodeling, and incident and prevalent AF in a community-based cohort. METHODS We analyzed left atrial function index (LAFI) of 1788 Framingham Offspring 8 participants. We quantified expression of 339 plasma microRNAs. We examined associations between microRNA levels with LAFI and prevalent and incident AF. We constructed pathway analysis of microRNAs' predicted gene targets to identify molecular processes involved in adverse atrial remodeling in AF. RESULTS The mean age of the participants was 66 ± 9 years, and 54% were women. Five percent of participants had prevalent AF at the initial examination and 9% (n = 157) developed AF over a median 8.6 years of follow-up (IQR 8.1-9.2 years). Plasma microRNAs were associated with LAFI (N = 73, p<0.0001). Six of these plasma microRNAs were significantly associated with incident AF, including 4 also associated with prevalent AF (microRNAs 106b, 26a-5p, 484, 20a-5p). These microRNAs are predicted to regulate genes involved in cardiac hypertrophy, inflammation, and myocardial fibrosis. CONCLUSIONS Circulating microRNAs 106b, 26a-5p, 484, 20a-5p are associated with atrial remodeling and AF.
Collapse
Affiliation(s)
- Aditya Vaze
- Division of Cardiology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Khanh-Van Tran
- Division of Cardiology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Kahraman Tanriverdi
- Division of Cardiology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Mayank Sardana
- Division of Cardiology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Darleen Lessard
- Division of Epidemiology of Chronic Diseases, Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - J. Kevin Donahue
- Division of Cardiology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Bruce Barton
- Biostatistics and Health Services Research, Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Gerard Aurigemma
- Division of Cardiology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Steven A. Lubitz
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Honghuang Lin
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, Massachusetts; Computational Biomedicine Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - George H. Nasr
- Department of Medicine, University of California Irvine, Orange, California, United States of America
| | - Amiya Mandapati
- Division of Cardiology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Emelia J. Benjamin
- Department of Medicine, and Department of Epidemiology, Boston University's and National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts; Section of Preventive Medicine and Epidemiology and Cardiovascular Medicine, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, United States of America
| | - Ramachandran S. Vasan
- Department of Medicine, and Department of Epidemiology, Boston University's and National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts; Section of Preventive Medicine and Epidemiology and Cardiovascular Medicine, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, United States of America
| | - Jane E. Freedman
- Division of Cardiology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - David D. McManus
- Division of Cardiology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
36
|
Genetics and Epigenetics of Atrial Fibrillation. Int J Mol Sci 2020; 21:ijms21165717. [PMID: 32784971 PMCID: PMC7460853 DOI: 10.3390/ijms21165717] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 12/13/2022] Open
Abstract
Atrial fibrillation (AF) is known to be the most common supraventricular arrhythmia affecting up to 1% of the general population. Its prevalence exponentially increases with age and could reach up to 8% in the elderly population. The management of AF is a complex issue that is addressed by extensive ongoing basic and clinical research. AF centers around different types of disturbances, including ion channel dysfunction, Ca2+-handling abnormalities, and structural remodeling. Genome-wide association studies (GWAS) have uncovered over 100 genetic loci associated with AF. Most of these loci point to ion channels, distinct cardiac-enriched transcription factors, as well as to other regulatory genes. Recently, the discovery of post-transcriptional regulatory mechanisms, involving non-coding RNAs (especially microRNAs), DNA methylation, and histone modification, has allowed to decipher how a normal heart develops and which modifications are involved in reshaping the processes leading to arrhythmias. This review aims to provide a current state of the field regarding the identification and functional characterization of AF-related epigenetic regulatory networks
Collapse
|
37
|
Ke ZP, Xu YJ, Wang ZS, Sun J. RNA sequencing profiling reveals key mRNAs and long noncoding RNAs in atrial fibrillation. J Cell Biochem 2020; 121:3752-3763. [PMID: 31680326 DOI: 10.1002/jcb.29504] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/08/2019] [Indexed: 01/24/2023]
Abstract
Long noncoding RNAs (lncRNAs) are an emerging class of RNA species that could participate in some critical pathways and disease pathogenesis. However, the underlying molecular mechanism of lncRNAs in atrial fibrillation (AF) is still not fully understood. In the present study, we analyzed RNA-seq data of paired left and right atrial appendages from five patients with AF and other five patients without AF. Based on the gene expression profiles of 20 samples, we found that a majority of genes were aberrantly expressed in both left and right atrial appendages of patients with AF. Similarly, the dysregulated pathways in the left and right atrial appendages of patients with AF also bore a close resemblance. Moreover, we predicted regulatory lncRNAs that regulated the expression of adjacent protein-coding genes (PCGs) or interacted with proteins. We identified that NPPA and its antisense RNA NPPA-AS1 may participate in the pathogenesis of AF by regulating the muscle contraction. We also identified that RP11 - 99E15.2 and RP3 - 523K23.2 could interact with proteins ITGB3 and HSF2, respectively. RP11 - 99E15.2 and RP3 - 523K23.2 may participate in the pathogenesis of AF via regulating the extracellular matrix binding and the transcription of HSF2 target genes, respectively. The close association of the lncRNA-interacting proteins with AF further demonstrated that these two lncRNAs were also associated with AF. In conclusion, we have identified key regulatory lncRNAs implicated in AF, which not only improves our understanding of the lncRNA-related molecular mechanism underlying AF but also provides computationally predicted regulatory lncRNAs for AF researchers.
Collapse
Affiliation(s)
- Zun-Ping Ke
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Ying-Jia Xu
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Zhang-Sheng Wang
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Jian Sun
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Clinical Research Unit, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
38
|
Clinical and Genetic Contributors to New-Onset Atrial Fibrillation in Critically Ill Adults. Crit Care Med 2020; 48:22-30. [PMID: 31599812 DOI: 10.1097/ccm.0000000000004034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES New-onset atrial fibrillation during critical illness is an independent risk factor for mortality. The ability to identify patients at high risk for new-onset atrial fibrillation is limited. We hypothesized that genetic susceptibility contributes to risk of new-onset atrial fibrillation in the ICU. DESIGN Retrospective sub-study of a prospective observational cohort study. SETTING Medical and general surgical ICUs in a tertiary academic medical center. PATIENTS One-thousand three-hundred sixty-nine critically ill patients admitted to the ICU for at least 2 days with no known history of atrial fibrillation who had DNA available for genotyping. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS We genotyped 21 single-nucleotide polymorphisms associated with atrial fibrillation in ambulatory studies using a Sequenom platform (San Diego, CA). We collected demographics, medical history, and development of new-onset atrial fibrillation during the first four days of ICU admission. New-onset atrial fibrillation occurred in 98 patients (7.2%) and was associated with age, male sex, coronary artery disease, and vasopressor use. Single-nucleotide polymorphisms associated with new-onset atrial fibrillation were rs3853445 (near PITX2, p = 0.0002), rs6838973 (near PITX2, p = 0.01), and rs12415501 (in NEURL, p = 0.03) on univariate testing. When controlling for clinical factors, rs3853445 (odds ratio, 0.47; 95% CI, 0.30-0.73; p = 0.001) and rs12415501 (odds ratio, 1.72; 95% CI, 1.27-2.59; p = 0.01) remained significantly associated with new-onset atrial fibrillation. The addition of genetic variables to clinical factors improved new-onset atrial fibrillation discrimination in a multivariable logistic regression model (C-statistic 0.82 vs 0.78; p = 0.0009). CONCLUSIONS We identified several single-nucleotide polymorphisms associated with new-onset atrial fibrillation in a large cohort of critically ill ICU patients, suggesting there is genetic susceptibility underlying this common clinical condition. This finding may provide new targets for future mechanistic studies and additional insight into the application of genomic information to identify patients at elevated risk for a common and important condition in the ICU.
Collapse
|
39
|
Huang X, Li Y, Zhang J, Wang X, Li Z, Li G. The molecular genetic basis of atrial fibrillation. Hum Genet 2020; 139:1485-1498. [PMID: 32617797 DOI: 10.1007/s00439-020-02203-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022]
Abstract
As the most common cardiac arrhythmia, atrial fibrillation (AF) is a major risk factor for stroke, heart failure, and premature death with considerable associated costs. However, no available treatment options have optimal benefit-harm profiles currently, reflecting an incomplete understanding of the biological mechanisms underlying this complex arrhythmia. Recently, molecular epidemiological studies, especially genome-wide association studies, have emphasized the substantial genetic component of AF etiology. A comprehensive mapping of the genetic underpinnings for AF can expand our knowledge of AF mechanism and further facilitate the process of locating novel therapeutics for AF. Here we provide a state-of-the-art review of the molecular genetics of AF incorporating evidence from linkage analysis and candidate gene, as well as genome-wide association studies of common variations and rare copy number variations; potential epigenetic modifications (e.g., DNA methylation, histone modification, and non-coding RNAs) are also involved. We also outline the challenges in mechanism investigation and potential future directions in this article.
Collapse
Affiliation(s)
- Xin Huang
- Center for Clinical Epidemiology and Methodology (CCEM), Guangdong Second Provincial General Hospital, 466 Newport Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China
| | - Yuhui Li
- Department of Cardiology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Junguo Zhang
- Center for Clinical Epidemiology and Methodology (CCEM), Guangdong Second Provincial General Hospital, 466 Newport Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China
| | - Xiaojie Wang
- Center for Clinical Epidemiology and Methodology (CCEM), Guangdong Second Provincial General Hospital, 466 Newport Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China
| | - Ziyi Li
- Center for Clinical Epidemiology and Methodology (CCEM), Guangdong Second Provincial General Hospital, 466 Newport Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China
| | - Guowei Li
- Center for Clinical Epidemiology and Methodology (CCEM), Guangdong Second Provincial General Hospital, 466 Newport Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China. .,Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University Hamilton, 1280 Main St West, Hamilton, ON, L8S 4L8, Canada.
| |
Collapse
|
40
|
Franco D, Aranega A, Dominguez JN. Non-coding RNAs and Atrial Fibrillation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:311-325. [PMID: 32285421 DOI: 10.1007/978-981-15-1671-9_19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Atrial fibrillation is the most frequent type of cardiac arrhythmia in humans, with an estimate incidence of 1-2% in the general population, rising up to 8-10% in the elderly. Cardiovascular risk factors such as diabetes, obesity, hypertension and hyperthyroidism can increase the occurrence of AF. The onset of AF triggers additional AF episodes, leading to structural and electrical remodeling of the diseased heart. Understanding the molecular bases of atrial fibrillation have greatly advance over the last decade demonstrating a pivotal role of distinct ion channels in AF pathophysiology. A new scenario has opened on the understanding of the molecular mechanisms underlying AF, with the discovery of non-coding RNAs and their wide implication in multiple disease states, including cardiac arrhythmogenic pathologies. microRNAs are small non-coding RNAs of 22-24 nucleotides that are capable of regulating gene expression by interacting with the mRNA transcript 3'UTRs and promoting mRNA degradation and/or protein translation blockage. Long non-coding RNAs are a more diverse group of non-coding RNAs, providing transcriptional and post-transcriptional roles and subclassified according to their functional properties. In this chapter we summarized current state-of-the-art knowledge on the functional of microRNAs and long non-coding RNAs as well as their cross-talk regulatory mechanisms in atrial fibrillation.
Collapse
Affiliation(s)
- Diego Franco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain.
| | - Amelia Aranega
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain
| | - Jorge N Dominguez
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain
| |
Collapse
|
41
|
van Ouwerkerk AF, Hall AW, Kadow ZA, Lazarevic S, Reyat JS, Tucker NR, Nadadur RD, Bosada FM, Bianchi V, Ellinor PT, Fabritz L, Martin J, de Laat W, Kirchhof P, Moskowitz I, Christoffels VM. Epigenetic and Transcriptional Networks Underlying Atrial Fibrillation. Circ Res 2020; 127:34-50. [PMID: 32717170 PMCID: PMC8315291 DOI: 10.1161/circresaha.120.316574] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Genome-wide association studies have uncovered over a 100 genetic loci associated with atrial fibrillation (AF), the most common arrhythmia. Many of the top AF-associated loci harbor key cardiac transcription factors, including PITX2, TBX5, PRRX1, and ZFHX3. Moreover, the vast majority of the AF-associated variants lie within noncoding regions of the genome where causal variants affect gene expression by altering the activity of transcription factors and the epigenetic state of chromatin. In this review, we discuss a transcriptional regulatory network model for AF defined by effector genes in Genome-wide association studies loci. We describe the current state of the field regarding the identification and function of AF-relevant gene regulatory networks, including variant regulatory elements, dose-sensitive transcription factor functionality, target genes, and epigenetic states. We illustrate how altered transcriptional networks may impact cardiomyocyte function and ionic currents that impact AF risk. Last, we identify the need for improved tools to identify and functionally test transcriptional components to define the links between genetic variation, epigenetic gene regulation, and atrial function.
Collapse
Affiliation(s)
- Antoinette F. van Ouwerkerk
- Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Amelia W. Hall
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Zachary A. Kadow
- Program in Developmental Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Sonja Lazarevic
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Jasmeet S. Reyat
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Nathan R. Tucker
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Masonic Medical Research Institute, Utica, NY, USA
| | - Rangarajan D. Nadadur
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Fernanda M. Bosada
- Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Valerio Bianchi
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Patrick T. Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Larissa Fabritz
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- SWBH and UHB NHS Trusts, Birmingham, UK
| | - Jim Martin
- Program in Developmental Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030
- Texas Heart Institute, Houston, Texas, 77030
| | - Wouter de Laat
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Paulus Kirchhof
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- SWBH and UHB NHS Trusts, Birmingham, UK
- University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Ivan Moskowitz
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Vincent M. Christoffels
- Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
42
|
Identification of microRNA biomarkers in serum of patients at different stages of atrial fibrillation. Heart Lung 2020; 49:902-908. [PMID: 32482362 DOI: 10.1016/j.hrtlng.2020.03.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/16/2020] [Accepted: 03/27/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) is a type of cardiac arrhythmia which is caused by irregular electrical activities in the atria. OBJECTIVE To identify serum microRNA (miRNA) biomarkers at three durations (duration since diagnosis of AF) of AF. METHODS This study included 14 patients with AF and 8 healthy subjects. The blood sample was collected from each patient at baseline (time of diagnosis) and 12-month and 24-month follow-up periods. The serum was used for miRNA sequencing. The differentially expressed miRNAs (DEMs) between the 3 AF and control groups were independently compared. The predicted target genes of DEMs were subjected to functional enrichment and protein-protein interaction network analyses. Additionally, the miRNA-target gene networks were constructed for the 3 AF groups and miRNA time series analysis was performed. The expression of several key miRNAs was verified by real-time quantitative polymerase chain reaction (qRT-PCR). RESULTS In total, 28, 22, and 24 DEMs were identified in the baseline, 12-month, and 24-month groups, respectively. miR-483-5p was the common DEM in the 3 AF groups. In the baseline and 12-month groups, the miR-200b-3p and miR-125b-5p target genes were significantly enriched in the Wnt signaling and several cancer-related pathways, respectively. In the 12-month group, the miR-34a-5p target genes were enriched in the cancer-related pathways. In the miRNA-target gene network, miR-34a-5p regulated the highest number of target genes. The time series analysis revealed that 7 miRNAs, which were downregulated in the control group, were upregulated in the AF groups. The qRT-PCR analysis revealed that the 24-month group exhibited a significant upregulation of miR-483-5p (p < 0.05), whereas the baseline group exhibited significant a downregulation of miR-125b-5p (p < 0.05). CONCLUSION In patients with AF, miR-125b-5p and miR-483-5p can be potential biomarkers of the baseline and 24-month periods, respectively.
Collapse
|
43
|
Wang L, Tang Q, Xu J, Li H, Yang T, Li L, Machon O, Hu T, Chen Y. The transcriptional regulator MEIS2 sets up the ground state for palatal osteogenesis in mice. J Biol Chem 2020; 295:5449-5460. [PMID: 32169905 PMCID: PMC7170518 DOI: 10.1074/jbc.ra120.012684] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 03/10/2020] [Indexed: 02/05/2023] Open
Abstract
Haploinsufficiency of Meis homeobox 2 (MEIS2), encoding a transcriptional regulator, is associated with human cleft palate, and Meis2 inactivation leads to abnormal palate development in mice, implicating MEIS2 functions in palate development. However, its functional mechanisms remain unknown. Here we observed widespread MEIS2 expression in the developing palate in mice. Wnt1Cre -mediated Meis2 inactivation in cranial neural crest cells led to a secondary palate cleft. Importantly, about half of the Wnt1Cre ;Meis2f/f mice exhibited a submucous cleft, providing a model for studying palatal bone formation and patterning. Consistent with complete absence of palatal bones, the results from integrative analyses of MEIS2 by ChIP sequencing, RNA-Seq, and an assay for transposase-accessible chromatin sequencing identified key osteogenic genes regulated directly by MEIS2, indicating that it plays a fundamental role in palatal osteogenesis. De novo motif analysis uncovered that the MEIS2-bound regions are highly enriched in binding motifs for several key osteogenic transcription factors, particularly short stature homeobox 2 (SHOX2). Comparative ChIP sequencing analyses revealed genome-wide co-occupancy of MEIS2 and SHOX2 in addition to their colocalization in the developing palate and physical interaction, suggesting that SHOX2 and MEIS2 functionally interact. However, although SHOX2 was required for proper palatal bone formation and was a direct downstream target of MEIS2, Shox2 overexpression failed to rescue the palatal bone defects in a Meis2-mutant background. These results, together with the fact that Meis2 expression is associated with high osteogenic potential and required for chromatin accessibility of osteogenic genes, support a vital function of MEIS2 in setting up a ground state for palatal osteogenesis.
Collapse
Affiliation(s)
- Linyan Wang
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China; Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Qinghuang Tang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Jue Xu
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118; West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Hua Li
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Tianfang Yang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Liwen Li
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Ondrej Machon
- Department of Developmental Biology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14200 Praha, Czech Republic
| | - Tao Hu
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - YiPing Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118.
| |
Collapse
|
44
|
Cxcr4 and Sdf-1 are critically involved in the formation of facial and non-somitic neck muscles. Sci Rep 2020; 10:5049. [PMID: 32193486 PMCID: PMC7081242 DOI: 10.1038/s41598-020-61960-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 03/05/2020] [Indexed: 01/22/2023] Open
Abstract
The present study shows that the CXCR4/SDF-1 axis regulates the migration of second branchial arch-derived muscles as well as non-somitic neck muscles. Cxcr4 is expressed by skeletal muscle progenitor cells in the second branchial arch (BA2). Muscles derived from the second branchial arch, but not from the first, fail to form in Cxcr4 mutants at embryonic days E13.5 and E14.5. Cxcr4 is also required for the development of non-somitic neck muscles. In Cxcr4 mutants, non-somitic neck muscle development is severely perturbed. In vivo experiments in chicken by means of loss-of-function approach based on the application of beads loaded with the CXCR4 inhibitor AMD3100 into the cranial paraxial mesoderm resulted in decreased expression of Tbx1 in the BA2. Furthermore, disrupting this chemokine signal at a later stage by implanting these beads into the BA2 caused a reduction in MyoR, Myf5 and MyoD expression. In contrast, gain-of-function experiments based on the implantation of SDF-1 beads into BA2 resulted in an attraction of myogenic progenitor cells, which was reflected in an expansion of the expression domain of these myogenic markers towards the SDF-1 source. Thus, Cxcr4 is required for the formation of the BA2 derived muscles and non-somitic neck muscles.
Collapse
|
45
|
Bai J, Lu Y, Lo A, Zhao J, Zhang H. PITX2 upregulation increases the risk of chronic atrial fibrillation in a dose-dependent manner by modulating IKs and ICaL -insights from human atrial modelling. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:191. [PMID: 32309338 PMCID: PMC7154416 DOI: 10.21037/atm.2020.01.90] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Functional analysis has shown that the paired-like homeodomain transcription factor 2 (PITX2) overexpression associated with atrial fibrillation (AF) leads to the slow delayed rectifier K+ current (IKs ) increase and the L-type Ca2+ current (ICaL ) reduction observed in isolated right atrial myocytes from chronic AF (CAF) patients. Through multiscale computational models, this study aimed to investigate the functional impact of the PITX2 overexpression on atrial electrical activity. METHODS The well-known Courtemanche-Ramirez-Nattel (CRN) model of human atrial action potentials (APs) was updated to incorporate experimental data on alterations in IKs and ICaL due to the PITX2 overexpression. These cell models for sinus rhythm (SR) and CAF were then incorporated into homogeneous multicellular one-dimensional (1D), two-dimensional (2D), and three-dimensional (3D) tissue models. The proarrhythmic effects of the PITX2 overexpression were quantified with ion current profiles, AP morphology, AP duration (APD) restitution, conduction velocity restitution (CVR), wavelength (WL), vulnerable window (VW) for unidirectional conduction block, and minimal substrate size required to induce re-entry. Dynamic behaviors of spiral waves were characterized by measuring lifespan (LS), tip patterns and dominant frequencies. RESULTS The IKs increase and the ICaL decrease arising from the PITX2 overexpression abbreviated APD and flattened APD restitution (APDR) curves in single cells. It reduced WL and increased CV at high excitation rates at the 1D tissue level. Although it had no effects on VW for initiating spiral waves, it decreased the minimal substrate size necessary to sustain re-entry. It also stabilized and accelerated spiral waves in 2D and 3D tissue models. CONCLUSIONS Electrical remodeling (IKs and ICaL ) due to the PITX2 overexpression increases susceptibility to AF due to increased tissue vulnerability, abbreviated APD, shortened WL and altered CV, which, in combination, facilitate initiation and maintenance of spiral waves.
Collapse
Affiliation(s)
- Jieyun Bai
- Department of Electronic Engineering, College of Information Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yaosheng Lu
- Department of Electronic Engineering, College of Information Science and Technology, Jinan University, Guangzhou 510632, China
| | - Andy Lo
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Jichao Zhao
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Henggui Zhang
- Biological Physics Group, School of Physics & Astronomy, University of Manchester, Manchester, UK
| |
Collapse
|
46
|
Xi J, Li QQ, Li BQ, Li N. miR‑155 inhibition represents a potential valuable regulator in mitigating myocardial hypoxia/reoxygenation injury through targeting BAG5 and MAPK/JNK signaling. Mol Med Rep 2020; 21:1011-1020. [PMID: 31922242 PMCID: PMC7003039 DOI: 10.3892/mmr.2020.10924] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 09/27/2019] [Indexed: 12/30/2022] Open
Abstract
Increasing evidence has indicated that miR-155 is closely associated with apoptosis, which may protect the myocardium and diminish the infarct area in myocardial ischemia reperfusion injury (IRI). In addition, studies have revealed that miR-155 serves a leading role in promoting fibroblast inflammation, cardiac dysfunction and other aspects of myocardial injury. The present study aimed to uncover the function and potential biological mechanism of miR-155 in myocardial IRI. The rat H9c2 myocardial cells was treated with hypoxia/reoxygenation (H/R) to simulate IRI in vitro. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to detect the expression levels of miR-155 mRNA. Cell Counting Kit-8 and flow cytometry assays and western blot analysis were applied to determine the biological behaviors of the H/R-treated cells. The association between miR-155 and BAG family molecular chaperone regulator 5 (BAG5) was predicted by bioinformatics software and was confirmed by dual luciferase assay. RT-qPCR and western blot analysis were used to analyze the expression of BAG5. The key proteins involved in mitogen-activated protein kinase (MAPK)/JNK signaling pathway were detected by western blot analysis. The data from the RT-qPCR assay indicated that the expression of miR-155 was markedly upregulated in the H/R model, and that downregulation of miR-155 may promote cell proliferation and inhibit cell apoptosis, and vice versa. BAG5, which was downregulated in the H/R model, was confirmed as a target of miR-155 and negatively modulated by miR-155. The key proteins involved in MAPK/JNK signaling, which were highly expressed in the H/R model, were suppressed by treatment with the miR-155 inhibitor, and overexpression of BAG5 promoted the protective effect of miR-155 inhibition on cell injury caused by H/R. In addition, the expression patterns of hypoxia-inducible factor 1-α and von Hippel-Lindau were altered following different treatments. Taken together, the data from the present study indicated that miR-155 inhibition represented a potential treatment strategy to improve myocardial H/R injury, which may be associated with targeting BAG5 and inhibition of the MAPK/JNK pathway.
Collapse
Affiliation(s)
- Jing Xi
- Department of Cardiology, Anqiu People's Hospital, Weifang, Shandong 262100, P.R. China
| | - Qiang-Qiang Li
- Department of Cardiology in Integrated Traditional Chinese and Western Medicine, Anqiu People's Hospital, Weifang, Shandong 262100, P.R. China
| | - Bing-Qiang Li
- Department of Cardiology, Anqiu People's Hospital, Weifang, Shandong 262100, P.R. China
| | - Ning Li
- Department of Cardiology, Anqiu People's Hospital, Weifang, Shandong 262100, P.R. China
| |
Collapse
|
47
|
Xu J, Wang L, Li H, Yang T, Zhang Y, Hu T, Huang Z, Chen Y. Shox2 regulates osteogenic differentiation and pattern formation during hard palate development in mice. J Biol Chem 2019; 294:18294-18305. [PMID: 31649032 DOI: 10.1074/jbc.ra119.008801] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 10/10/2019] [Indexed: 02/05/2023] Open
Abstract
During mammalian palatogenesis, cranial neural crest-derived mesenchymal cells undergo osteogenic differentiation and form the hard palate, which is divided into palatine process of the maxilla and the palatine. However, it remains unknown whether these bony structures originate from the same cell lineage and how the hard palate is patterned at the molecular level. Using mice, here we report that deficiency in Shox2 (short stature homeobox 2), a transcriptional regulator whose expression is restricted to the anterior palatal mesenchyme, leads to a defective palatine process of the maxilla but does not affect the palatine. Shox2 overexpression in palatal mesenchyme resulted in a hyperplastic palatine process of the maxilla and a hypoplastic palatine. RNA sequencing and assay for transposase-accessible chromatin-sequencing analyses revealed that Shox2 controls the expression of pattern specification and skeletogenic genes associated with accessible chromatin in the anterior palate. This highlighted a lineage-autonomous function of Shox2 in patterning and osteogenesis of the hard palate. H3K27ac ChIP-Seq and transient transgenic enhancer assays revealed that Shox2 binds distal-acting cis-regulatory elements in an anterior palate-specific manner. Our results suggest that the palatine process of the maxilla and palatine arise from different cell lineages and differ in ossification mechanisms. Shox2 evidently controls osteogenesis of a cell lineage and contributes to the palatine process of the maxilla by interacting with distal cis-regulatory elements to regulate skeletogenic gene expression and to pattern the hard palate. Genome-wide Shox2 occupancy in the developing palate may provide a marker for identifying active anterior palate-specific gene enhancers.
Collapse
Affiliation(s)
- Jue Xu
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan, China; West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041 Sichuan, China; Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Linyan Wang
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan, China; Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Hua Li
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118; Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neuro Biology, College of Life Sciences, Fujian Normal University, Fuzhou, 350117 Fujian, China
| | - Tianfang Yang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Yanding Zhang
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neuro Biology, College of Life Sciences, Fujian Normal University, Fuzhou, 350117 Fujian, China
| | - Tao Hu
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan, China.
| | - Zhen Huang
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neuro Biology, College of Life Sciences, Fujian Normal University, Fuzhou, 350117 Fujian, China.
| | - YiPing Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118.
| |
Collapse
|
48
|
Yin L, Liu MX, Wang FY, Wang X, Tang YH, Zhao QY, Wang T, Chen YT, Huang CX. Transcription Factor prrx1 Promotes Brown Adipose-Derived Stem Cells Differentiation to Sinus Node-Like Cells. DNA Cell Biol 2019; 38:1313-1322. [PMID: 31545082 DOI: 10.1089/dna.2019.4998] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
This study investigated whether overexpression of paired-related homeobox 1 (prrx1) can successfully induce differentiation of brown adipose-derived stem cells (BADSCs) into sinus node-like cells. The experiments were performed in two groups: adenovirus-green fluorescent protein (Ad-GFP) group and Ad-prrx1 group. After 5-7 days of adenoviral transfection, the expression levels of sinus node cell-associated pacing protein (hyperpolarization-activated cyclic nucleotide-gated potassium channel 4 [HCN4]) and ion channel (calcium channel, voltage-dependent, T type, alpha 1G subunit [Cacna1g]), as well as transcription factors (T-box 18 [TBX18], insulin gene enhancer binding protein 1 [ISL-1], paired-like homeodomain transcription factor 2 [pitx2], short stature homeobox 2 [shox2]), were detected by western blot and reverse transcription-quantitative polymerase chain reaction. Immunofluorescence assay was carried out to detect whether prrx1 was coexpressed with HCN4, TBX18, and ISL-1. Finally, whole-cell patch-clamp technique was used to record pacing current hyperpolarization-activated inward current (If). The isolated cells were CD90+, CD29+, and CD45-, indicating that pure BADSCs were successfully isolated. After 5-7 days of Ad transfection into cells, the mRNA levels and protein levels of pacing-related factors (TBX18, ISL-1, HCN4, shox2, and Cacna1g) in Ad-prrx1 group were significantly higher than those in Ad-GFP group. However, the expression level of pitx2 was decreased. Immunofluorescence analysis showed that prrx1 was coexpressed with TBX18, ISL-1, and HCN4 in the Ad-prrx1 group, which did not appear in the Ad-GFP group. Whole-cell patch clamps were able to record the If current in the experimental group rather than in the Ad-GFP group. Overexpression of prrx1 can successfully induce sinus node-like cells.
Collapse
Affiliation(s)
- Lin Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Ming-Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Feng-Yuan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Yan-Hong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Qing-Yan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Teng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Yu-Ting Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Cong-Xin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| |
Collapse
|
49
|
Abstract
The rate and rhythm of heart muscle contractions are coordinated by the cardiac conduction system (CCS), a generic term for a collection of different specialized muscular tissues within the heart. The CCS components initiate the electrical impulse at the sinoatrial node, propagate it from atria to ventricles via the atrioventricular node and bundle branches, and distribute it to the ventricular muscle mass via the Purkinje fibre network. The CCS thereby controls the rate and rhythm of alternating contractions of the atria and ventricles. CCS function is well conserved across vertebrates from fish to mammals, although particular specialized aspects of CCS function are found only in endotherms (mammals and birds). The development and homeostasis of the CCS involves transcriptional and regulatory networks that act in an embryonic-stage-dependent, tissue-dependent, and dose-dependent manner. This Review describes emerging data from animal studies, stem cell models, and genome-wide association studies that have provided novel insights into the transcriptional networks underlying CCS formation and function. How these insights can be applied to develop disease models and therapies is also discussed.
Collapse
|
50
|
Ren J, Han P, Ma X, Farah EN, Bloomekatz J, Zeng XXI, Zhang R, Swim MM, Witty AD, Knight HG, Deshpande R, Xu W, Yelon D, Chen S, Chi NC. Canonical Wnt5b Signaling Directs Outlying Nkx2.5+ Mesoderm into Pacemaker Cardiomyocytes. Dev Cell 2019; 50:729-743.e5. [PMID: 31402282 DOI: 10.1016/j.devcel.2019.07.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 05/01/2019] [Accepted: 07/11/2019] [Indexed: 12/21/2022]
Abstract
Pacemaker cardiomyocytes that create the sinoatrial node are essential for the initiation and maintenance of proper heart rhythm. However, illuminating developmental cues that direct their differentiation has remained particularly challenging due to the unclear cellular origins of these specialized cardiomyocytes. By discovering the origins of pacemaker cardiomyocytes, we reveal an evolutionarily conserved Wnt signaling mechanism that coordinates gene regulatory changes directing mesoderm cell fate decisions, which lead to the differentiation of pacemaker cardiomyocytes. We show that in zebrafish, pacemaker cardiomyocytes derive from a subset of Nkx2.5+ mesoderm that responds to canonical Wnt5b signaling to initiate the cardiac pacemaker program, including activation of pacemaker cell differentiation transcription factors Isl1 and Tbx18 and silencing of Nkx2.5. Moreover, applying these developmental findings to human pluripotent stem cells (hPSCs) notably results in the creation of hPSC-pacemaker cardiomyocytes, which successfully pace three-dimensional bioprinted hPSC-cardiomyocytes, thus providing potential strategies for biological cardiac pacemaker therapy.
Collapse
Affiliation(s)
- Jie Ren
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Peidong Han
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Xuanyi Ma
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Elie N Farah
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Joshua Bloomekatz
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093, USA; Department of Biology, University of Mississippi, Oxford, MS 38677, USA
| | - Xin-Xin I Zeng
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ruilin Zhang
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Megan M Swim
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Alec D Witty
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hannah G Knight
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Rima Deshpande
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Weizhe Xu
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Deborah Yelon
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shaochen Chen
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA; Institute for Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Neil C Chi
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093, USA; Institute for Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|