1
|
Sirish P, Diloretto DA, Thai PN, Chiamvimonvat N. The Critical Roles of Proteostasis and Endoplasmic Reticulum Stress in Atrial Fibrillation. Front Physiol 2022; 12:793171. [PMID: 35058801 PMCID: PMC8764384 DOI: 10.3389/fphys.2021.793171] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/08/2021] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) remains the most common arrhythmia seen clinically. The incidence of AF is increasing due to the aging population. AF is associated with a significant increase in morbidity and mortality, yet current treatment paradigms have proven largely inadequate. Therefore, there is an urgent need to develop new effective therapeutic strategies for AF. The endoplasmic reticulum (ER) in the heart plays critical roles in the regulation of excitation-contraction coupling and cardiac function. Perturbation in the ER homeostasis due to intrinsic and extrinsic factors, such as inflammation, oxidative stress, and ischemia, leads to ER stress that has been linked to multiple conditions including diabetes mellitus, neurodegeneration, cancer, heart disease, and cardiac arrhythmias. Recent studies have documented the critical roles of ER stress in the pathophysiological basis of AF. Using an animal model of chronic pressure overload, we demonstrate a significant increase in ER stress in atrial tissues. Moreover, we demonstrate that treatment with a small molecule inhibitor to inhibit the soluble epoxide hydrolase enzyme in the arachidonic acid metabolism significantly reduces ER stress as well as atrial electrical and structural remodeling. The current review article will attempt to provide a perspective on our recent understandings and current knowledge gaps on the critical roles of proteostasis and ER stress in AF progression.
Collapse
Affiliation(s)
- Padmini Sirish
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, United States.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA, United States
| | - Daphne A Diloretto
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, United States
| | - Phung N Thai
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, United States.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA, United States
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, United States.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA, United States.,Department of Pharmacology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
2
|
Gähwiler EKN, Motta SE, Martin M, Nugraha B, Hoerstrup SP, Emmert MY. Human iPSCs and Genome Editing Technologies for Precision Cardiovascular Tissue Engineering. Front Cell Dev Biol 2021; 9:639699. [PMID: 34262897 PMCID: PMC8273765 DOI: 10.3389/fcell.2021.639699] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/31/2021] [Indexed: 12/12/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) originate from the reprogramming of adult somatic cells using four Yamanaka transcription factors. Since their discovery, the stem cell (SC) field achieved significant milestones and opened several gateways in the area of disease modeling, drug discovery, and regenerative medicine. In parallel, the emergence of clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (CRISPR-Cas9) revolutionized the field of genome engineering, allowing the generation of genetically modified cell lines and achieving a precise genome recombination or random insertions/deletions, usefully translated for wider applications. Cardiovascular diseases represent a constantly increasing societal concern, with limited understanding of the underlying cellular and molecular mechanisms. The ability of iPSCs to differentiate into multiple cell types combined with CRISPR-Cas9 technology could enable the systematic investigation of pathophysiological mechanisms or drug screening for potential therapeutics. Furthermore, these technologies can provide a cellular platform for cardiovascular tissue engineering (TE) approaches by modulating the expression or inhibition of targeted proteins, thereby creating the possibility to engineer new cell lines and/or fine-tune biomimetic scaffolds. This review will focus on the application of iPSCs, CRISPR-Cas9, and a combination thereof to the field of cardiovascular TE. In particular, the clinical translatability of such technologies will be discussed ranging from disease modeling to drug screening and TE applications.
Collapse
Affiliation(s)
- Eric K. N. Gähwiler
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Sarah E. Motta
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Wyss Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Marcy Martin
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, United States
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA, United States
| | - Bramasta Nugraha
- Molecular Parasitology Lab, Institute of Parasitology, University of Zurich, Zurich, Switzerland
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Simon P. Hoerstrup
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Wyss Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Maximilian Y. Emmert
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Wyss Zurich, University and ETH Zurich, Zurich, Switzerland
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
| |
Collapse
|
3
|
Liu SH, Lo LW, Chou YH, Lin WL, Tsai TY, Cheng WH, Yamada S, Chen SA. Renal denervation prevents myocardial structural remodeling and arrhythmogenicity in a chronic kidney disease rabbit model. Heart Rhythm 2021; 18:1596-1604. [PMID: 33992732 DOI: 10.1016/j.hrthm.2021.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 04/30/2021] [Accepted: 05/09/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND The electrophysiological (EP) effects and safety of renal artery denervation (RDN) in chronic kidney disease (CKD) are unclear. OBJECTIVE The purpose of this study was to investigate the arrhythmogenicity of RDN in a rabbit model of CKD. METHODS Eighteen New Zealand white rabbits were randomized to control (n = 6), CKD (n = 6), and CKD-RDN (n = 6) groups. A 5/6 nephrectomy was selected for the CKD model. RDN was applied in the CKD-RDN group. All rabbits underwent cardiac EP studies for evaluation. Immunohistochemistry, myocardial fibrosis, and renal catecholamine levels were evaluated. RESULTS The CKD group (34.8% ± 9.2%) had a significantly higher ventricular arrhythmia (VA) inducibility than the control (8.6% ± 3.8%; P <.01) and CKD-RDN (19.5% ± 6.3%; P = .01) groups. In the CKD-RDN group, ventricular fibrosis was significantly decreased compared to the CKD group (7.4% ± 2.0 % vs 10.4% ± 3.7%; P = .02). Sympathetic innervation in the CKD group was significantly increased compared to the control and CKD-RDN groups [left ventricle: 4.1 ± 1.8 vs 0.8 ± 0.5 (102 μm2/mm2), P <.01; 4.1 ± 1.8 vs 0.9± 0.6 (102 μm2/mm2), P <.01; right ventricle: 3.6 ± 1.0 vs 1.0 ± 0.4 (102 μm2/mm2), P <.01; 3.6 ± 1.0 vs 1.0 ± 0.5 (102 μm2/mm2), P <.01]. CONCLUSION Neuromodulation by RDN demonstrated protective effects with less structural and electrical remodeling, leading to attenuated VAs. In a rabbit model of CKD, RDN plays a therapeutic role by lowering the risk of VA caused by autonomic dysfunction.
Collapse
Affiliation(s)
- Shin-Huei Liu
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine and Cardiovascular Research Institute, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Li-Wei Lo
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine and Cardiovascular Research Institute, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Yu-Hui Chou
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine and Cardiovascular Research Institute, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Lun Lin
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine and Cardiovascular Research Institute, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tseng-Ying Tsai
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine and Cardiovascular Research Institute, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wen-Han Cheng
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine and Cardiovascular Research Institute, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shinya Yamada
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shih-Ann Chen
- Institute of Clinical Medicine and Cardiovascular Research Institute, National Yang Ming Chiao Tung University, Taipei, Taiwan; Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan
| |
Collapse
|
4
|
Arrhythmia Mechanisms in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. J Cardiovasc Pharmacol 2020; 77:300-316. [PMID: 33323698 DOI: 10.1097/fjc.0000000000000972] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/08/2020] [Indexed: 12/30/2022]
Abstract
ABSTRACT Despite major efforts by clinicians and researchers, cardiac arrhythmia remains a leading cause of morbidity and mortality in the world. Experimental work has relied on combining high-throughput strategies with standard molecular and electrophysiological studies, which are, to a great extent, based on the use of animal models. Because this poses major challenges for translation, the progress in the development of novel antiarrhythmic agents and clinical care has been mostly disappointing. Recently, the advent of human induced pluripotent stem cell-derived cardiomyocytes has opened new avenues for both basic cardiac research and drug discovery; now, there is an unlimited source of cardiomyocytes of human origin, both from healthy individuals and patients with cardiac diseases. Understanding arrhythmic mechanisms is one of the main use cases of human induced pluripotent stem cell-derived cardiomyocytes, in addition to pharmacological cardiotoxicity and efficacy testing, in vitro disease modeling, developing patient-specific models and personalized drugs, and regenerative medicine. Here, we review the advances that the human induced pluripotent stem cell-derived-based modeling systems have brought so far regarding the understanding of both arrhythmogenic triggers and substrates, while also briefly speculating about the possibilities in the future.
Collapse
|
5
|
Santoso F, Farhan A, Castillo AL, Malhotra N, Saputra F, Kurnia KA, Chen KHC, Huang JC, Chen JR, Hsiao CD. An Overview of Methods for Cardiac Rhythm Detection in Zebrafish. Biomedicines 2020; 8:E329. [PMID: 32899676 PMCID: PMC7554775 DOI: 10.3390/biomedicines8090329] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/29/2020] [Accepted: 09/01/2020] [Indexed: 12/17/2022] Open
Abstract
The heart is the most important muscular organ of the cardiovascular system, which pumps blood and circulates, supplying oxygen and nutrients to peripheral tissues. Zebrafish have been widely explored in cardiotoxicity research. For example, the zebrafish embryo has been used as a human heart model due to its body transparency, surviving several days without circulation, and facilitating mutant identification to recapitulate human diseases. On the other hand, adult zebrafish can exhibit the amazing regenerative heart muscle capacity, while adult mammalian hearts lack this potential. This review paper offers a brief description of the major methodologies used to detect zebrafish cardiac rhythm at both embryonic and adult stages. The dynamic pixel change method was mostly performed for the embryonic stage. Other techniques, such as kymography, laser confocal microscopy, artificial intelligence, and electrocardiography (ECG) have also been applied to study heartbeat in zebrafish embryos. Nevertheless, ECG is widely used for heartbeat detection in adult zebrafish since ECG waveforms' similarity between zebrafish and humans is prominent. High-frequency ultrasound imaging (echocardiography) and modern electronic sensor tag also have been proposed. Despite the fact that each method has its benefits and limitations, it is proved that zebrafish have become a promising animal model for human cardiovascular disease, drug pharmaceutical, and toxicological research. Using those tools, we conclude that zebrafish behaviors as an excellent small animal model to perform real-time monitoring for the developmental heart process with transparent body appearance, to conduct the in vivo cardiovascular performance and gene function assays, as well as to perform high-throughput/high content drug screening.
Collapse
Affiliation(s)
- Fiorency Santoso
- Master Program in Nanotechnology, Chung Yuan Christian University, Chung-Li 320314, Taiwan;
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 320314, Taiwan; (F.S.); (K.A.K.)
| | - Ali Farhan
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Punjab 38000, Pakistan;
| | - Agnes L. Castillo
- Faculty of Pharmacy, The Graduate School and Research Center for the Natural and Applied Sciences, University of Santo Tomas, Manila 1008, Philippines;
| | - Nemi Malhotra
- Department of Biomedical Engineering, Chung Yuan Christian University, Chung-Li 320314, Taiwan;
| | - Ferry Saputra
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 320314, Taiwan; (F.S.); (K.A.K.)
| | - Kevin Adi Kurnia
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 320314, Taiwan; (F.S.); (K.A.K.)
| | - Kelvin H.-C. Chen
- Department of Applied Chemistry, National Pingtung University, Pingtung 900391, Taiwan;
| | - Jong-Chin Huang
- Department of Applied Chemistry, National Pingtung University, Pingtung 900391, Taiwan;
| | - Jung-Ren Chen
- Department of Biological Science & Technology College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Chung-Der Hsiao
- Master Program in Nanotechnology, Chung Yuan Christian University, Chung-Li 320314, Taiwan;
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 320314, Taiwan; (F.S.); (K.A.K.)
- Center of Nanotechnology, Chung Yuan Christian University, Chung-Li 320314, Taiwan
| |
Collapse
|
6
|
Abstract
The term phenotype is so commonly used that we often assume that we each mean the same thing. The general definition, the set of observable characteristics of an individual resulting from the interaction of their genotype with the environment, is often left to the eye of the beholder. Whether applied to the multiple levels of biological phenomena or the intact human being, our ability to characterize, classify, and analyze phenotype has been limited by measurement deficits, computing limitations, and a culture that avoids the generalizable. With the advent of modern technology, there is the potential for a revolution in phenotyping, which incorporates old and new in structured ways to dramatically advance basic understanding of biology and behavior and to lead to major improvements in clinical care and public health. This revolution in how we think about phenotypes will require a radical change in the scale at which biomedicine operates with significant changes in the unit of action, which will have far-reaching implications for how care, translation, and discovery are implemented.
Collapse
Affiliation(s)
- Calum A MacRae
- From the One Brave Idea (C.A.M., R.M.C.).,Cardiovascular Medicine Division and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.A.M.)
| | - Robert M Califf
- From the One Brave Idea (C.A.M., R.M.C.).,Verily Life Sciences (R.M.C.).,Google Health, South San Francisco and Mountain View, CA (R.M.C.)
| |
Collapse
|
7
|
Hulsmans M, Aguirre AD, Bonner MD, Bapat A, Cremer S, Iwamoto Y, King KR, Swirski FK, Milan DJ, Weissleder R, Nahrendorf M. A Miniaturized, Programmable Pacemaker for Long-Term Studies in the Mouse. Circ Res 2019; 123:1208-1219. [PMID: 30571465 DOI: 10.1161/circresaha.118.313429] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Cardiac pacing is a critical technology for the treatment of arrhythmia and heart failure. The impact of specific pacing strategies on myocardial function is an area of intense research and high clinical significance. Mouse models have proven extremely useful for probing mechanisms of heart disease, but there is currently no reliable technology for long-term pacing in the mouse. OBJECTIVE We sought to develop a device for long-term pacing studies in mice. We evaluated the device for (1) treating third-degree atrioventricular block after macrophage depletion, (2) ventricular pacing-induced cardiomyopathy, and (3) high-rate atrial pacing. METHODS AND RESULTS We developed a mouse pacemaker by refashioning a 26 mm×6.7 mm clinical device powered by a miniaturized, highly efficient battery. The electrode was fitted with a single flexible lead, and custom software extended the pacing rate to up to 1200 bpm. The wirelessly programmable device was implanted in the dorsal subcutaneous space of 39 mice. The tunneled lead was passed through a left thoracotomy incision and attached to the epicardial surface of the apex (for ventricular pacing) or the left atrium (for atrial pacing). Mice tolerated the implantation and both long-term atrial and ventricular pacing over weeks. We then validated the pacemaker's suitability for the treatment of atrioventricular block after macrophage depletion in Cd11b DTR mice. Ventricular pacing increased the heart rate from 313±59 to 550 bpm ( P<0.05). In addition, we characterized tachypacing-induced cardiomyopathy in mice. Four weeks of ventricular pacing resulted in reduced left ventricular function, fibrosis, and an increased number of cardiac leukocytes and endothelial activation. Finally, we demonstrated the feasibility of chronic atrial pacing at 1200 bpm. CONCLUSIONS Long-term pacing with a fully implantable, programmable, and battery-powered device enables previously impossible investigations of arrhythmia and heart failure in the mouse.
Collapse
Affiliation(s)
- Maarten Hulsmans
- From the Department of Radiology, Center for Systems Biology (M.H., A.D.A., S.C., Y.I., K.R.K., F.K.S., R.W., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Aaron D Aguirre
- From the Department of Radiology, Center for Systems Biology (M.H., A.D.A., S.C., Y.I., K.R.K., F.K.S., R.W., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston.,Cardiology Division (A.D.A., D.J.M.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Matthew D Bonner
- Cardiac Rhythm and Heart Failure, Medtronic PLC, Mounds View, MN (M.D.B.)
| | - Aneesh Bapat
- Cardiovascular Research Center (A.B., D.J.M., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston.,Cardiology Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (A.B.)
| | - Sebastian Cremer
- From the Department of Radiology, Center for Systems Biology (M.H., A.D.A., S.C., Y.I., K.R.K., F.K.S., R.W., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Yoshiko Iwamoto
- From the Department of Radiology, Center for Systems Biology (M.H., A.D.A., S.C., Y.I., K.R.K., F.K.S., R.W., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Kevin R King
- From the Department of Radiology, Center for Systems Biology (M.H., A.D.A., S.C., Y.I., K.R.K., F.K.S., R.W., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston.,Department of Bioengineering, Jacobs School of Engineering (K.R.K.), University of California San Diego, La Jolla.,Department of Medicine, Cardiology Division (K.R.K.), University of California San Diego, La Jolla
| | - Filip K Swirski
- From the Department of Radiology, Center for Systems Biology (M.H., A.D.A., S.C., Y.I., K.R.K., F.K.S., R.W., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - David J Milan
- Cardiology Division (A.D.A., D.J.M.), Massachusetts General Hospital and Harvard Medical School, Boston.,Cardiovascular Research Center (A.B., D.J.M., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston.,Program in Population and Medical Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA (D.J.M.)
| | - Ralph Weissleder
- From the Department of Radiology, Center for Systems Biology (M.H., A.D.A., S.C., Y.I., K.R.K., F.K.S., R.W., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston.,Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Matthias Nahrendorf
- From the Department of Radiology, Center for Systems Biology (M.H., A.D.A., S.C., Y.I., K.R.K., F.K.S., R.W., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston.,Cardiovascular Research Center (A.B., D.J.M., M.N.), Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
8
|
MacRae CA. Closing the 'phenotype gap' in precision medicine: improving what we measure to understand complex disease mechanisms. Mamm Genome 2019; 30:201-211. [PMID: 31428846 DOI: 10.1007/s00335-019-09810-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 06/30/2019] [Indexed: 10/26/2022]
Abstract
The central concept underlying precision medicine is a mechanistic understanding of each disease and its response to therapy sufficient to direct a specific intervention. To execute on this vision requires parsing incompletely defined disease syndromes into discrete mechanistic subsets and developing interventions to precisely address each of these etiologically distinct entities. This will require substantial adjustment of traditional paradigms which have tended to aggregate high-level phenotypes with very different etiologies. In the current environment, where diagnoses are not mechanistic, drug development has become so expensive that it is now impractical to imagine the cost-effective creation of new interventions for many prevalent chronic conditions. The vision of precision medicine also argues for a much more seamless integration of research and development with clinical care, where shared taxonomies will enable every clinical interaction to inform our collective understanding of disease mechanisms and drug responses. Ideally, this would be executed in ways that drive real-time and real-world discovery, innovation, translation, and implementation. Only in oncology, where at least some of the biology is accessible through surgical excision of the diseased tissue or liquid biopsy, has "co-clinical" modeling proven feasible. In most common germline disorders, while genetics often reveal the causal mutations, there still remain substantial barriers to efficient disease modeling. Aggregation of similar disorders under single diagnostic labels has directly contributed to the paucity of etiologic and mechanistic understanding by directly reducing the resolution of any subsequent studies. Existing clinical phenotypes are typically anatomic, physiologic, or histologic, and result in a substantial mismatch in information content between the phenomes in humans or in animal 'models' and the variation in the genome. This lack of one-to-one mapping of discrete mechanisms between disease and animal models causes a failure of translation and is one form of 'phenotype gap.' In this review, we will focus on the origins of the phenotyping deficit and approaches that may be considered to bridge the gap, creating shared taxonomies between human diseases and relevant models, using cardiovascular examples.
Collapse
Affiliation(s)
- Calum A MacRae
- Cardiovascular Medicine, Genetics and Network Medicine Divisions, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Hale 7016, 75 Francis Street, Boston, MA, 02115, USA.
| |
Collapse
|
9
|
Pabon MA, Manocha K, Cheung JW, Lo JC. Linking Arrhythmias and Adipocytes: Insights, Mechanisms, and Future Directions. Front Physiol 2018; 9:1752. [PMID: 30568603 PMCID: PMC6290087 DOI: 10.3389/fphys.2018.01752] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/20/2018] [Indexed: 01/14/2023] Open
Abstract
Obesity and atrial fibrillation have risen to epidemic levels worldwide and may continue to grow over the next decades. Emerging evidence suggests that obesity promotes atrial and ventricular arrhythmias. This has led to trials employing various strategies with the ultimate goal of decreasing the atrial arrhythmic burden in obese patients. The effectiveness of these interventions remains to be determined. Obesity is defined by the expansion of adipose mass, making adipocytes a prime candidate to mediate the pro-arrhythmogenic effects of obesity. The molecular mechanisms linking obesity and adipocytes to increased arrhythmogenicity in both the atria and ventricles remain poorly understood. In this focused review, we highlight areas of potential molecular interplay between adipocytes and cardiomyocytes. The effects of adipocytes may be direct, local or remote. Direct effect refers to adipocyte or fatty infiltration of the atrial and ventricular myocardium itself, possibly causing increased dispersion of normal myocardial electrical signals and fibrotic substrate of adipocytes that promote reentry or adipocytes serving as a direct source of aberrant signals. Local effects may originate from nearby adipose depots, specifically epicardial adipose tissue (EAT) and pericardial adipose tissue, which may play a role in the secretion of adipokines and chemokines that can incite inflammation given the direct contact and disrupt the conduction system. Adipocytes can also have a remote effect on the myocardium arising from their systemic secretion of adipokines, cytokines and metabolites. These factors may lead to mitochondrial dysfunction, oxidative stress, autophagy, mitophagy, autonomic dysfunction, and cardiomyocyte death to ultimately produce a pro-arrhythmogenic state. By better understanding the molecular mechanisms connecting dysfunctional adipocytes and arrhythmias, novel therapies may be developed to sever the link between obesity and arrhythmias.
Collapse
Affiliation(s)
- Maria A Pabon
- Joan and Sanford I. Weill, Department of Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - Kevin Manocha
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - Jim W Cheung
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - James C Lo
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States.,Metabolic Health Center, Weill Cornell Medicine, New York, NY, United States.,Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
10
|
Scardigli M, Müllenbroich C, Margoni E, Cannazzaro S, Crocini C, Ferrantini C, Coppini R, Yan P, Loew LM, Campione M, Bocchi L, Giulietti D, Cerbai E, Poggesi C, Bub G, Pavone FS, Sacconi L. Real-time optical manipulation of cardiac conduction in intact hearts. J Physiol 2018; 596:3841-3858. [PMID: 29989169 PMCID: PMC6117584 DOI: 10.1113/jp276283] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/05/2018] [Indexed: 11/28/2022] Open
Abstract
Key points Although optogenetics has clearly demonstrated the feasibility of cardiac manipulation, current optical stimulation strategies lack the capability to react acutely to ongoing cardiac wave dynamics. Here, we developed an all‐optical platform to monitor and control electrical activity in real‐time. The methodology was applied to restore normal electrical activity after atrioventricular block and to manipulate the intraventricular propagation of the electrical wavefront. The closed‐loop approach was also applied to simulate a re‐entrant circuit across the ventricle. The development of this innovative optical methodology provides the first proof‐of‐concept that a real‐time all‐optical stimulation can control cardiac rhythm in normal and abnormal conditions.
Abstract Optogenetics has provided new insights in cardiovascular research, leading to new methods for cardiac pacing, resynchronization therapy and cardioversion. Although these interventions have clearly demonstrated the feasibility of cardiac manipulation, current optical stimulation strategies do not take into account cardiac wave dynamics in real time. Here, we developed an all‐optical platform complemented by integrated, newly developed software to monitor and control electrical activity in intact mouse hearts. The system combined a wide‐field mesoscope with a digital projector for optogenetic activation. Cardiac functionality could be manipulated either in free‐run mode with submillisecond temporal resolution or in a closed‐loop fashion: a tailored hardware and software platform allowed real‐time intervention capable of reacting within 2 ms. The methodology was applied to restore normal electrical activity after atrioventricular block, by triggering the ventricle in response to optically mapped atrial activity with appropriate timing. Real‐time intraventricular manipulation of the propagating electrical wavefront was also demonstrated, opening the prospect for real‐time resynchronization therapy and cardiac defibrillation. Furthermore, the closed‐loop approach was applied to simulate a re‐entrant circuit across the ventricle demonstrating the capability of our system to manipulate heart conduction with high versatility even in arrhythmogenic conditions. The development of this innovative optical methodology provides the first proof‐of‐concept that a real‐time optically based stimulation can control cardiac rhythm in normal and abnormal conditions, promising a new approach for the investigation of the (patho)physiology of the heart. Although optogenetics has clearly demonstrated the feasibility of cardiac manipulation, current optical stimulation strategies lack the capability to react acutely to ongoing cardiac wave dynamics. Here, we developed an all‐optical platform to monitor and control electrical activity in real‐time. The methodology was applied to restore normal electrical activity after atrioventricular block and to manipulate the intraventricular propagation of the electrical wavefront. The closed‐loop approach was also applied to simulate a re‐entrant circuit across the ventricle. The development of this innovative optical methodology provides the first proof‐of‐concept that a real‐time all‐optical stimulation can control cardiac rhythm in normal and abnormal conditions.
Collapse
Affiliation(s)
- M Scardigli
- European Laboratory for Non-Linear Spectroscopy, Florence, 50019, Italy.,National Institute of Optics, National Research Council, Florence, 50125, Italy
| | - C Müllenbroich
- European Laboratory for Non-Linear Spectroscopy, Florence, 50019, Italy.,National Institute of Optics, National Research Council, Florence, 50125, Italy
| | - E Margoni
- European Laboratory for Non-Linear Spectroscopy, Florence, 50019, Italy.,Department of Physics, University of Pisa, Pisa, 56127, Italy
| | - S Cannazzaro
- European Laboratory for Non-Linear Spectroscopy, Florence, 50019, Italy.,National Institute of Optics, National Research Council, Florence, 50125, Italy
| | - C Crocini
- European Laboratory for Non-Linear Spectroscopy, Florence, 50019, Italy.,National Institute of Optics, National Research Council, Florence, 50125, Italy
| | - C Ferrantini
- Division of Physiology, Department of Experimental and Clinical Medicine, University of Florence, Florence, 50134, Italy
| | - R Coppini
- Division of Pharmacology, Department 'NeuroFarBa', University of Florence, Florence, 50139, Italy
| | - P Yan
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - L M Loew
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - M Campione
- Neuroscience Institute, National Research Council, Padova, 35121, Italy.,Department of Biomedical Sciences, Univercity ot Padua, Padua, 35121, Italy
| | - L Bocchi
- European Laboratory for Non-Linear Spectroscopy, Florence, 50019, Italy.,Department of Information Engineering, University of Florence, Via S. Marta 3, Florence, 50139, Italy
| | - D Giulietti
- National Institute of Optics, National Research Council, Florence, 50125, Italy.,Department of Physics, University of Pisa, Pisa, 56127, Italy
| | - E Cerbai
- Division of Pharmacology, Department 'NeuroFarBa', University of Florence, Florence, 50139, Italy
| | - C Poggesi
- Division of Physiology, Department of Experimental and Clinical Medicine, University of Florence, Florence, 50134, Italy
| | - G Bub
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - F S Pavone
- European Laboratory for Non-Linear Spectroscopy, Florence, 50019, Italy.,National Institute of Optics, National Research Council, Florence, 50125, Italy.,Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, 50019, Italy
| | - L Sacconi
- European Laboratory for Non-Linear Spectroscopy, Florence, 50019, Italy.,National Institute of Optics, National Research Council, Florence, 50125, Italy
| |
Collapse
|
11
|
Kurganov NA, Blinova EV, Semeleva EV, Gromova IA, Blinov DS, Novikov AV, Mashkova JN, Vasilkina OV. 2-aminoaethanesulfonic acid compounds possess protective property in reperfusion-induced heart jnjury. RESEARCH RESULTS IN PHARMACOLOGY 2018. [DOI: 10.3897/rrpharmacology.4.27435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The study aim was to explore pharmacological effects of 2-aminoaethansulfonic acid compounds in reperfusion-induced heart injury.Materials and methods.The study was performed on rats and dogs of both sexes, isolated rats’ hearts. Two compounds of 2-aminoethanesulfonic acid, magnesium-containing (LBK-527) and phenylacetamide-containing (LKhT-317) were investigated. Antiarrhythmic effects of the compounds were studied in coronary artery reperfusion 7, 30 and 120 min after acute myocardial ischemia modeling. The ability of the substances to limit the volume of reperfusion injury was investigated by differential indicator method. The influence of substances on the intensity of free radical processes in the myocardium, as well as the metabolic profile of coronary venous blood during reperfusion, was studied. Hemodynamic effects of the substances were studied during in vivo experiments, as well as on an isolated heart.Results and Discussion.The compounds effectively prevent cardiac arrhythmias generation caused by myocardial reperfusion after 7, 30 and 120 minutes of ischemia. Prophylactic intravenous administration of LHT-317 and LBK-527 at higher therapeutic doses limit the size of rats’ heart necrosis zone after occlusion-reperfusion syndrome develops, prevent reperfusion-induced excessive activation of free-radical processes in rat myocardium, activate the antiradical activity of the heart tissues, and optimize [O2] and [CO2] in coronary venous sinus blood of dogs. Cardioprotective effect of the compounds manifests in preserving myocardium contractile function, maintaing BP and stabilizing heart chronotropic function.Conclusions.The study analysis shows that 2-aminoethanesulfonic acid compounds have cardioprotective effect in reperfusion syndrome.
Collapse
|
12
|
Lenning M, Fortunato J, Le T, Clark I, Sherpa A, Yi S, Hofsteen P, Thamilarasu G, Yang J, Xu X, Han HD, Hsiai TK, Cao H. Real-Time Monitoring and Analysis of Zebrafish Electrocardiogram with Anomaly Detection. SENSORS 2017; 18:s18010061. [PMID: 29283402 PMCID: PMC5796315 DOI: 10.3390/s18010061] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 12/21/2017] [Accepted: 12/24/2017] [Indexed: 12/13/2022]
Abstract
Heart disease is the leading cause of mortality in the U.S. with approximately 610,000 people dying every year. Effective therapies for many cardiac diseases are lacking, largely due to an incomplete understanding of their genetic basis and underlying molecular mechanisms. Zebrafish (Danio rerio) are an excellent model system for studying heart disease as they enable a forward genetic approach to tackle this unmet medical need. In recent years, our team has been employing electrocardiogram (ECG) as an efficient tool to study the zebrafish heart along with conventional approaches, such as immunohistochemistry, DNA and protein analyses. We have overcome various challenges in the small size and aquatic environment of zebrafish in order to obtain ECG signals with favorable signal-to-noise ratio (SNR), and high spatial and temporal resolution. In this paper, we highlight our recent efforts in zebrafish ECG acquisition with a cost-effective simplified microelectrode array (MEA) membrane providing multi-channel recording, a novel multi-chamber apparatus for simultaneous screening, and a LabVIEW program to facilitate recording and processing. We also demonstrate the use of machine learning-based programs to recognize specific ECG patterns, yielding promising results with our current limited amount of zebrafish data. Our solutions hold promise to carry out numerous studies of heart diseases, drug screening, stem cell-based therapy validation, and regenerative medicine.
Collapse
Affiliation(s)
- Michael Lenning
- School of STEM, University of Washington Bothell, Bothell, WA 98011, USA.
| | - Joseph Fortunato
- School of STEM, University of Washington Bothell, Bothell, WA 98011, USA.
| | - Tai Le
- School of STEM, University of Washington Bothell, Bothell, WA 98011, USA.
| | - Isaac Clark
- School of Medicine, University of Washington, Seattle, WA 98109, USA.
| | - Ang Sherpa
- School of STEM, University of Washington Bothell, Bothell, WA 98011, USA.
| | - Soyeon Yi
- School of STEM, University of Washington Bothell, Bothell, WA 98011, USA.
| | - Peter Hofsteen
- School of Medicine, University of Washington, Seattle, WA 98109, USA.
| | | | - Jingchun Yang
- Department of Biochemistry and Molecular Biology, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA.
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA.
| | - Huy-Dung Han
- School of Electronics and Telecommunications, Hanoi University of Science and Technology, Hanoi, Vietnam.
| | - Tzung K Hsiai
- School of Medicine, University of California Los Angeles, Los Angeles, CA 90073, USA.
| | - Hung Cao
- School of STEM, University of Washington Bothell, Bothell, WA 98011, USA.
- School of Medicine, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
13
|
Sala L, Bellin M, Mummery CL. Integrating cardiomyocytes from human pluripotent stem cells in safety pharmacology: has the time come? Br J Pharmacol 2017; 174:3749-3765. [PMID: 27641943 PMCID: PMC5647193 DOI: 10.1111/bph.13577] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 07/27/2016] [Accepted: 08/11/2016] [Indexed: 12/20/2022] Open
Abstract
Cardiotoxicity is a severe side effect of drugs that induce structural or electrophysiological changes in heart muscle cells. As a result, the heart undergoes failure and potentially lethal arrhythmias. It is still a major reason for drug failure in preclinical and clinical phases of drug discovery. Current methods for predicting cardiotoxicity are based on guidelines that combine electrophysiological analysis of cell lines expressing ion channels ectopically in vitro with animal models and clinical trials. Although no new cases of drugs linked to lethal arrhythmias have been reported since the introduction of these guidelines in 2005, their limited predictive power likely means that potentially valuable drugs may not reach clinical practice. Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are now emerging as potentially more predictive alternatives, particularly for the early phases of preclinical research. However, these cells are phenotypically immature and culture and assay methods not standardized, which could be a hurdle to the development of predictive computational models and their implementation into the drug discovery pipeline, in contrast to the ambitions of the comprehensive pro-arrhythmia in vitro assay (CiPA) initiative. Here, we review present and future preclinical cardiotoxicity screening and suggest possible hPSC-CM-based strategies that may help to move the field forward. Coordinated efforts by basic scientists, companies and hPSC banks to standardize experimental conditions for generating reliable and reproducible safety indices will be helpful not only for cardiotoxicity prediction but also for precision medicine. LINKED ARTICLES This article is part of a themed section on New Insights into Cardiotoxicity Caused by Chemotherapeutic Agents. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.21/issuetoc.
Collapse
Affiliation(s)
- Luca Sala
- Department of Anatomy and EmbryologyLeiden University Medical CenterLeidenZAThe Netherlands
| | - Milena Bellin
- Department of Anatomy and EmbryologyLeiden University Medical CenterLeidenZAThe Netherlands
| | - Christine L Mummery
- Department of Anatomy and EmbryologyLeiden University Medical CenterLeidenZAThe Netherlands
- Department of Applied Stem Cell TechnologiesUniversity of TwenteEnschedeThe Netherlands
| |
Collapse
|
14
|
Kithcart A, MacRae CA. Using Zebrafish for High-Throughput Screening of Novel Cardiovascular Drugs. JACC Basic Transl Sci 2017; 2:1-12. [PMID: 30167552 PMCID: PMC6113531 DOI: 10.1016/j.jacbts.2017.01.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 01/17/2017] [Accepted: 01/17/2017] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases remain a major challenge for modern drug discovery. The diseases are chronic, complex, and the result of sophisticated interactions between genetics and environment involving multiple cell types and a host of systemic factors. The clinical events are often abrupt, and the diseases may be asymptomatic until a highly morbid event. Target selection is often based on limited information, and though highly specific agents are often identified in screening, their final efficacy is often compromised by unanticipated systemic responses, a narrow therapeutic index, or substantial toxicities. Our understanding of complexity of cardiovascular disease has grown dramatically over the past 2 decades, and the range of potential disease mechanisms now includes pathways previously thought only tangentially involved in cardiac or vascular disease. Despite these insights, the majority of active cardiovascular agents derive from a remarkably small number of classes of agents and target a very limited number of pathways. These agents have often been used initially for particular indications and then discovered serendipitously to have efficacy in other cardiac disorders or in a manner unrelated to their original mechanism of action. In this review, the rationale for in vivo screening is described, and the utility of the zebrafish for this approach and for complementary work in functional genomics is discussed. Current limitations of the model in this setting and the need for careful validation in new disease areas are also described. An overview is provided of the complex mechanisms underlying most clinical cardiovascular diseases, and insight is offered into the limits of single downstream pathways as drug targets. The zebrafish is introduced as a model organism, in particular for cardiovascular biology. Potential approaches to overcoming the hurdles to drug discovery in the face of complex biology are discussed, including in vivo screening of zebrafish genetic disease models.
Collapse
Affiliation(s)
- Aaron Kithcart
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,The Broad Institute of MIT and Harvard, Harvard Stem Cell Institute, Boston, Massachusetts
| | - Calum A MacRae
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,The Broad Institute of MIT and Harvard, Harvard Stem Cell Institute, Boston, Massachusetts
| |
Collapse
|
15
|
Kuroda Y, Yuasa S, Watanabe Y, Ito S, Egashira T, Seki T, Hattori T, Ohno S, Kodaira M, Suzuki T, Hashimoto H, Okata S, Tanaka A, Aizawa Y, Murata M, Aiba T, Makita N, Furukawa T, Shimizu W, Kodama I, Ogawa S, Kokubun N, Horigome H, Horie M, Kamiya K, Fukuda K. Flecainide ameliorates arrhythmogenicity through NCX flux in Andersen-Tawil syndrome-iPS cell-derived cardiomyocytes. Biochem Biophys Rep 2017; 9:245-256. [PMID: 28956012 PMCID: PMC5614591 DOI: 10.1016/j.bbrep.2017.01.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 12/09/2016] [Accepted: 01/10/2017] [Indexed: 12/15/2022] Open
Abstract
Andersen-Tawil syndrome (ATS) is a rare inherited channelopathy. The cardiac phenotype in ATS is typified by a prominent U wave and ventricular arrhythmia. An effective treatment for this disease remains to be established. We reprogrammed somatic cells from three ATS patients to generate induced pluripotent stem cells (iPSCs). Multi-electrode arrays (MEAs) were used to record extracellular electrograms of iPSC-derived cardiomyocytes, revealing strong arrhythmic events in the ATS-iPSC-derived cardiomyocytes. Ca2+ imaging of cells loaded with the Ca2+ indicator Fluo-4 enabled us to examine intracellular Ca2+ handling properties, and we found a significantly higher incidence of irregular Ca2+ release in the ATS-iPSC-derived cardiomyocytes than in control-iPSC-derived cardiomyocytes. Drug testing using ATS-iPSC-derived cardiomyocytes further revealed that antiarrhythmic agent, flecainide, but not the sodium channel blocker, pilsicainide, significantly suppressed these irregular Ca2+ release and arrhythmic events, suggesting that flecainide's effect in these cardiac cells was not via sodium channels blocking. A reverse-mode Na+/Ca2+exchanger (NCX) inhibitor, KB-R7943, was also found to suppress the irregular Ca2+ release, and whole-cell voltage clamping of isolated guinea-pig cardiac ventricular myocytes confirmed that flecainide could directly affect the NCX current (INCX). ATS-iPSC-derived cardiomyocytes recapitulate abnormal electrophysiological phenotypes and flecainide suppresses the arrhythmic events through the modulation of INCX. iPS cells are generated from three patients with ATS. ATS-iPS cell-derived cardiomyocytes show abnormal electrophysiological phenotypes. Flecainide suppresses abnormal electrophysiological phenotypes in ATS-iPS cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Yusuke Kuroda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan.,Department of Cardiovascular Research, Research Institute of Environmental Medicine, Nagoya University, Aichi, Japan.,Department of Cardiology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Shinsuke Yuasa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Yasuhide Watanabe
- Division of Pharmacological Science, Department of Health Science, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Shogo Ito
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Toru Egashira
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Tomohisa Seki
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Tetsuhisa Hattori
- Department of Cardiovascular Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Seiko Ohno
- Department of Cardiovascular Medicine, Shiga University of Medical Science, Shiga, Japan.,Center for Epidemiologic Research in Asia, Shiga University of Medical Science, Shiga, Japan
| | - Masaki Kodaira
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Tomoyuki Suzuki
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan.,Department of Cardiovascular Research, Research Institute of Environmental Medicine, Nagoya University, Aichi, Japan.,Department of Cardiology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Hisayuki Hashimoto
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Shinichiro Okata
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Atsushi Tanaka
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshiyasu Aizawa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Mitsushige Murata
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan.,Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takeshi Aiba
- Division of Arrhythmia and Electrophysiology, Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Naomasa Makita
- Department of Molecular Pathophysiology-1, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tetsushi Furukawa
- Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School, Tokyo, Japan
| | - Itsuo Kodama
- Department of Cardiovascular Research, Research Institute of Environmental Medicine, Nagoya University, Aichi, Japan
| | - Satoshi Ogawa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Norito Kokubun
- Department of Neurology, Dokkyo Medical University, Tochigi, Japan
| | - Hitoshi Horigome
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Minoru Horie
- Department of Cardiovascular Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Kaichiro Kamiya
- Department of Cardiovascular Research, Research Institute of Environmental Medicine, Nagoya University, Aichi, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
16
|
Matsa E, Ahrens JH, Wu JC. Human Induced Pluripotent Stem Cells as a Platform for Personalized and Precision Cardiovascular Medicine. Physiol Rev 2016; 96:1093-126. [PMID: 27335446 PMCID: PMC6345246 DOI: 10.1152/physrev.00036.2015] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) have revolutionized the field of human disease modeling, with an enormous potential to serve as paradigm shifting platforms for preclinical trials, personalized clinical diagnosis, and drug treatment. In this review, we describe how hiPSCs could transition cardiac healthcare away from simple disease diagnosis to prediction and prevention, bridging the gap between basic and clinical research to bring the best science to every patient.
Collapse
Affiliation(s)
- Elena Matsa
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, and Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - John H Ahrens
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, and Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, and Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
17
|
Lozekoot PWJ, de Jong MMJ, Parise O, Matteucci F, Lucà F, Kumar N, Bani D, Czapla J, Pison L, Crijns HJGM, Maessen JG, La Meir M, Gelsomino S. The ABLA-BOX: An in Vitro Module of Hybrid Atrial Fibrillation Ablation. INNOVATIONS-TECHNOLOGY AND TECHNIQUES IN CARDIOTHORACIC AND VASCULAR SURGERY 2016. [DOI: 10.1177/155698451601100308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Pieter W. J. Lozekoot
- Cardiology and Cardiothoracic Department, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Monique M. J. de Jong
- Cardiology and Cardiothoracic Department, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Orlando Parise
- Cardiology and Cardiothoracic Department, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Francesco Matteucci
- Cardiology and Cardiothoracic Department, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Fabiana Lucà
- Cardiology and Cardiothoracic Department, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Narendra Kumar
- Cardiology and Cardiothoracic Department, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Daniele Bani
- Department of Clinical and Experimental Medicine, Careggi University Hospital-University of Florence, Florence, Italy
| | - Jens Czapla
- Department of Cardiothoracic Surgery, University Hospital, Brussels, Belgium
| | - Laurent Pison
- Cardiology and Cardiothoracic Department, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Harry J. G. M. Crijns
- Cardiology and Cardiothoracic Department, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Jos G. Maessen
- Cardiology and Cardiothoracic Department, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Mark La Meir
- Cardiology and Cardiothoracic Department, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Cardiothoracic Surgery, University Hospital, Brussels, Belgium
| | - Sandro Gelsomino
- Cardiology and Cardiothoracic Department, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Clinical and Experimental Medicine, Careggi University Hospital-University of Florence, Florence, Italy
| |
Collapse
|
18
|
The ABLA-BOX: An in Vitro Module of Hybrid Atrial Fibrillation Ablation. INNOVATIONS-TECHNOLOGY AND TECHNIQUES IN CARDIOTHORACIC AND VASCULAR SURGERY 2016; 11:201-9. [PMID: 27355130 DOI: 10.1097/imi.0000000000000256] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Objective We present the first testing study on the ABLA-BOX, a new in vitro module of hybrid atrial fibrillation ablation. Methods ABLA-BOX consists of two chambers that mimic the epicardial and endocardial sides of the heart. The septum between chambers provides catheter access on both sites of the cardiac tissue. A circuit, filled with freshly obtained porcine blood, including a pump, an oxygenator, and a heating device, circulate the blood inside the system. Left atrial fresh tissue is mounted on a tissue holder and magnetically fixed. Epicardial and endocardial catheters are fixed on the catheter holders and blocked with the locker knob. The system allows control of ablation force, flow rate, temperature, and flow pattern. Results Epicardial contact force of 100 g and endocardial force of 30 g resulted in larger lesion volumes (P < 0.001), areas (P < 0.001), and lesion diameters (P = 0.03 and P = 0.008), than the combination of 100/20 g. In addition, with a flow rate of 5 L/min, lesion volumes (P = 0.02), areas (P < 0.001), and diameters (both, P < 0.001) were significantly larger in comparison with those of 3 L/min. Furthermore, dimensions (both, P < 0.001), volume (P < 0.001), and area (P < 0.001) of the lesions at a circulating blood temperature of 38.0°C were larger than with a lower blood temperature (36.0°C). Finally, ablations made under stable flow pattern resulted in greater lesion diameters P = 0.04 and P = 0.03) as well as larger volumes (P = 0.02) and areas (P = 0.03) than under turbulent-like flow reproduced with the system rotor set to 400 rpm. Conclusions The ABLA-BOX allowed easy hybrid ablation with different setups, which can provide cardiologists and cardiac surgeons with reliable and more valuable insights.
Collapse
|
19
|
Lee J, Cao H, Kang BJ, Jen N, Yu F, Lee CA, Fei P, Park J, Bohlool S, Lash-Rosenberg L, Shung KK, Hsiai TK. Hemodynamics and ventricular function in a zebrafish model of injury and repair. Zebrafish 2015; 11:447-54. [PMID: 25237983 DOI: 10.1089/zeb.2014.1016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Myocardial infarction results in scar tissue and irreversible loss of ventricular function. Unlike humans, zebrafish has the capacity to remove scar tissue after injury. To assess ventricular function during repair, we synchronized microelectrocardiogram (μECG) signals with a high-frequency ultrasound pulsed-wave (PW) Doppler to interrogate cardiac hemodynamics. μECG signals allowed for identification of PW Doppler signals for passive (early [E]-wave velocity) and active ventricular filling (atrial [A]-wave velocity) during diastole. The A wave (9.0±1.2 cm·s(-1)) is greater than the E wave (1.1±0.4 cm·s(-1)), resulting in an E/A ratio <1 (0.12±0.05, n=6). In response to cryocauterization to the ventricular epicardium, the E-wave velocity increased, accompanied by a rise in the E/A ratio at 3 days postcryocauterization (dpc) (0.55±0.13, n=6, p<0.001 vs. sham). The E waves normalize toward the baseline, along with a reduction in the E/A ratio at 35 dpc (0.36±0.06, n=6, p<0.001 vs. sham) and 65 dpc (0.2±0.16, n=6, p<0.001 vs. sham). In zebrafish, E/A<1 at baseline is observed, suggesting the distinct two-chamber system in which the pressure gradient across the atrioventricular valve is higher compared with the ventriculobulbar valve. The initial rise and subsequent normalization of E/A ratios support recovery in the ventricular diastolic function.
Collapse
Affiliation(s)
- Juhyun Lee
- 1 Division of Cardiology, Department of Medicine, University of California , Los Angeles, Los Angeles, California
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Hassinen M, Haverinen J, Vornanen M. Molecular basis and drug sensitivity of the delayed rectifier (IKr) in the fish heart. Comp Biochem Physiol C Toxicol Pharmacol 2015. [PMID: 26215639 DOI: 10.1016/j.cbpc.2015.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Fishes are increasingly used as models for human cardiac diseases, creating a need for a better understanding of the molecular basis of fish cardiac ion currents. To this end we cloned KCNH6 channel of the crucian carp (Carassius carassius) that produces the rapid component of the delayed rectifier K(+) current (IKr), the main repolarising current of the fish heart. KCNH6 (ccErg2) was the main isoform of the Kv11 potassium channel family with relative transcript levels of 98.9% and 99.6% in crucian carp atrium and ventricle, respectively. KCNH2 (ccErg1), an orthologue to human cardiac Erg (Herg) channel, was only slightly expressed in the crucian carp heart. The native atrial IKr and the cloned ccErg2 were inhibited by similar concentrations of verapamil, terfenadine and KB-R7943 (P>0.05), while the atrial IKr was about an order of magnitude more sensitive to E-4031 than ccErg2 (P<0.05) suggesting that some accessory β-subunits may be involved. Sensitivity of the crucian carp atrial IKr to E-4031, terfenadine and KB-R7943 was similar to what has been reported for the Herg channel. In contrast, the sensitivity of the crucian carp IKr to verapamil was approximately 30 times higher than the previously reported values for the Herg current. In conclusion, the cardiac IKr is produced by non-orthologous gene products in fish (Erg2) and mammalian hearts (Erg1) and some marked differences exist in drug sensitivity between fish and mammalian Erg1/2 which need to be taken into account when using fish heart as a model for human heart.
Collapse
Affiliation(s)
- Minna Hassinen
- University of Eastern Finland, Department of Biology, P.O. Box 111, 80101 Joensuu, Finland.
| | - Jaakko Haverinen
- University of Eastern Finland, Department of Biology, P.O. Box 111, 80101 Joensuu, Finland
| | - Matti Vornanen
- University of Eastern Finland, Department of Biology, P.O. Box 111, 80101 Joensuu, Finland
| |
Collapse
|
21
|
Nguyen MN, Kiriazis H, Ruggiero D, Gao XM, Su Y, Jian A, Han LP, McMullen JR, Du XJ. Spontaneous ventricular tachyarrhythmias in β2-adrenoceptor transgenic mice in relation to cardiac interstitial fibrosis. Am J Physiol Heart Circ Physiol 2015; 309:H946-57. [PMID: 26116714 DOI: 10.1152/ajpheart.00405.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 06/26/2015] [Indexed: 12/21/2022]
Abstract
Myocardial fibrosis is regarded as a pivotal proarrhythmic substrate, but there have been no comprehensive studies showing a correlation between the severity of fibrosis and ventricular tachyarrhythmias (VTAs). Our purpose was to document this relationship in a transgenic (TG) strain of mice with fibrotic cardiomyopathy. TG mice with cardiac overexpression of β2-adrenoceptors (β2-AR mice) and non-TG (NTG) littermates were studied at 4-12 mo of age. VTA was quantified by ECG telemetry. The effect of pharmacological blockade of β2-ARs on VTA was examined. Myocardial collagen content was determined by hydroxyproline assay. NTG and TG mice displayed circadian variation in heart rate, which was higher in TG mice than in NTG mice (P <0.05). Frequent spontaneous ventricular ectopic beats (VEBs) and ventricular tachycardia (VT) were prominent in TG mice but not present in NTG mice. The frequency of VEB and VT episodes in TG mice increased with age (P < 0.01). Ventricular collagen content was greater in TG mice than in NTG mice (P <0.001) and correlated with age (r = 0.71, P < 0.01). The number of VEBs or VT episodes correlated with age (r = 0.83 and r = 0.73) and the content of total or cross-linked collagen (r = 0.62∼0.66, all P <0.01). While having no effect in younger β2-TG mice, β2-AR blockade reduced the frequency of VTA in old β2-TG mice with more severe fibrosis. In conclusion, β2-TG mice exhibit interstitial fibrosis and spontaneous onset of VTA, becoming more severe with aging. The extent of cardiac fibrosis is a major determinant for both the frequency of VTA and proarrhythmic action of β2-AR activation.
Collapse
Affiliation(s)
- My-Nhan Nguyen
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Helen Kiriazis
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Diego Ruggiero
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; University of Milan, Milan, Italy
| | - Xiao-Ming Gao
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Yidan Su
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Anne Jian
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Li-Ping Han
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; WenZhou Medical University, WenZhou, China; and
| | - Julie R McMullen
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Physiology, Monash University, Melbourne, Australia
| | - Xiao-Jun Du
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; Central Clinical School, Monash University, Melbourne, Victoria, Australia;
| |
Collapse
|
22
|
Cao H, Kang BJ, Lee CA, Shung KK, Hsiai TK. Electrical and Mechanical Strategies to Enable Cardiac Repair and Regeneration. IEEE Rev Biomed Eng 2015; 8:114-24. [PMID: 25974948 DOI: 10.1109/rbme.2015.2431681] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Inadequate replacement of lost ventricular myocardium from myocardial infarction leads to heart failure. Investigating the regenerative capacity of mammalian hearts represents an emerging direction for tissue engineering and cell-based therapy. Recent advances in stem cells hold promise to restore cardiac functions. However, embryonic or induced pluripotent stem cell-derived cardiomyocytes lack functional phenotypes of the native myocardium, and transplanted tissues are not fully integrated for synchronized electrical and mechanical coupling with the host. In this context, this review highlights the mechanical and electrical strategies to promote cardiomyocyte maturation and integration, and to assess the functional phenotypes of regenerating myocardium. Simultaneous microelectrocardiogram and high-frequency ultrasound techniques will also be introduced to assess electrical and mechanical coupling for small animal models of heart regeneration.
Collapse
|
23
|
Cao H, Yu F, Zhao Y, Zhang X, Tai J, Lee J, Darehzereshki A, Bersohn M, Lien CL, Chi NC, Tai YC, Hsiai TK. Wearable multi-channel microelectrode membranes for elucidating electrophysiological phenotypes of injured myocardium. Integr Biol (Camb) 2015; 6:789-95. [PMID: 24945366 DOI: 10.1039/c4ib00052h] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Understanding the regenerative capacity of small vertebrate models has provided new insights into the plasticity of injured myocardium. Here, we demonstrate the application of flexible microelectrode arrays (MEAs) in elucidating electrophysiological phenotypes of zebrafish and neonatal mouse models of heart regeneration. The 4-electrode MEA membranes were designed to detect electrical signals in the aquatic environment. They were micro-fabricated to adhere to the non-planar body surface of zebrafish and neonatal mice. The acquired signals were processed to display an electrocardiogram (ECG) with high signal-to-noise-ratios, and were validated via the use of conventional micro-needle electrodes. The 4-channel MEA provided signal stability and spatial resolution, revealing the site-specific electrical injury currents such as ST-depression in response to ventricular cryo-injury. Thus, our polymer-based and wearable MEA membranes provided electrophysiological insights into long-term conduction phenotypes for small vertebral models of heart injury and regeneration with a translational implication for monitoring cardiac patients.
Collapse
Affiliation(s)
- Hung Cao
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Ozcan C, Battaglia E, Young R, Suzuki G. LKB1 knockout mouse develops spontaneous atrial fibrillation and provides mechanistic insights into human disease process. J Am Heart Assoc 2015; 4:e001733. [PMID: 25773299 PMCID: PMC4392447 DOI: 10.1161/jaha.114.001733] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 01/14/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) is a complex disease process, and the molecular mechanisms underlying initiation and progression of the disease are unclear. Consequently, AF has been difficult to model. In this study, we have presented a novel transgenic mouse model of AF that mimics human disease and characterized the mechanisms of atrial electroanatomical remodeling in the genesis of AF. METHODS AND RESULTS Cardiac-specific liver kinase B1 (LKB1) knockout (KO) mice were generated, and 47% aged 4 weeks and 95% aged 12 weeks developed spontaneous AF from sinus rhythm by demonstrating paroxysmal and persistent stages of the disease. Electrocardiographic characteristics of sinus rhythm were similar in KO and wild-type mice. Atrioventricular block and atrial flutter were common in KO mice. Heart rate was slower with persistent AF. In parallel with AF, KO mice developed progressive biatrial enlargement with inflammation, heterogeneous fibrosis, and loss of cardiomyocyte population with apoptosis and necrosis. Atrial tissue was infiltrated with inflammatory cells. C-reactive protein, interleukin 6, and tumor necrosis factor α were significantly elevated in serum. KO atria demonstrated elevated reactive oxygen species and decreased AMP-activated protein kinase activity. Cardiomyocyte and myofibrillar ultrastructure were disrupted. Intercellular matrix and gap junction were interrupted. Connexins 40 and 43 were reduced. Persistent AF caused left ventricular dysfunction and heart failure. Survival and exercise capacity were worse in KO mice. CONCLUSIONS LKB1 KO mice develop spontaneous AF from sinus rhythm and progress into persistent AF by replicating the human AF disease process. Progressive inflammatory atrial cardiomyopathy is the genesis of AF, through mechanistic electrical and structural remodeling.
Collapse
Affiliation(s)
- Cevher Ozcan
- Division of Cardiovascular Medicine, Department of Medicine, Clinical & Translational Research Center, University at Buffalo School of Medicine and Biomedical Sciences, Buffalo, NY (C.O., E.B., R.Y., G.S.)
- Section of Cardiology, Department of Medicine, University of Chicago, IL (C.O.)
| | - Emily Battaglia
- Division of Cardiovascular Medicine, Department of Medicine, Clinical & Translational Research Center, University at Buffalo School of Medicine and Biomedical Sciences, Buffalo, NY (C.O., E.B., R.Y., G.S.)
| | - Rebeccah Young
- Division of Cardiovascular Medicine, Department of Medicine, Clinical & Translational Research Center, University at Buffalo School of Medicine and Biomedical Sciences, Buffalo, NY (C.O., E.B., R.Y., G.S.)
| | - Gen Suzuki
- Division of Cardiovascular Medicine, Department of Medicine, Clinical & Translational Research Center, University at Buffalo School of Medicine and Biomedical Sciences, Buffalo, NY (C.O., E.B., R.Y., G.S.)
| |
Collapse
|
25
|
Kurhanewicz N, McIntosh-Kastrinsky R, Tong H, Walsh L, Farraj AK, Hazari MS. Ozone co-exposure modifies cardiac responses to fine and ultrafine ambient particulate matter in mice: concordance of electrocardiogram and mechanical responses. Part Fibre Toxicol 2014; 11:54. [PMID: 25318591 PMCID: PMC4203862 DOI: 10.1186/s12989-014-0054-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/02/2014] [Indexed: 12/19/2022] Open
Abstract
Background Studies have shown a relationship between air pollution and increased risk of cardiovascular morbidity and mortality. Due to the complexity of ambient air pollution composition, recent studies have examined the effects of co-exposure, particularly particulate matter (PM) and gas, to determine whether pollutant interactions alter (e.g. synergistically, antagonistically) the health response. This study examines the independent effects of fine (FCAPs) and ultrafine (UFCAPs) concentrated ambient particles on cardiac function, and determine the impact of ozone (O3) co-exposure on the response. We hypothesized that UFCAPs would cause greater decrement in mechanical function and electrical dysfunction than FCAPs, and that O3 co-exposure would enhance the effects of both particle-types. Methods Conscious/unrestrained radiotelemetered mice were exposed once whole-body to either 190 μg/m3 FCAPs or 140 μg/m3 UFCAPs with/without 0.3 ppm O3; separate groups were exposed to either filtered air (FA) or O3 alone. Heart rate (HR) and electrocardiogram (ECG) were recorded continuously before, during and after exposure, and cardiac mechanical function was assessed using a Langendorff perfusion preparation 24 hrs post-exposure. Results FCAPs alone caused a significant decrease in baseline left ventricular developed pressure (LVDP) and contractility, whereas UFCAPs did not; neither FCAPs nor UFCAPs alone caused any ECG changes. O3 co-exposure with FCAPs caused a significant decrease in heart rate variability when compared to FA but also blocked the decrement in cardiac function. On the other hand, O3 co-exposure with UFCAPs significantly increased QRS-interval, QTc and non-conducted P-wave arrhythmias, and decreased LVDP, rate of contractility and relaxation when compared to controls. Conclusions These data suggest that particle size and gaseous interactions may play a role in cardiac function decrements one day after exposure. Although FCAPs + O3 only altered autonomic balance, UFCAPs + O3 appeared to be more serious by increasing cardiac arrhythmias and causing mechanical decrements. As such, O3 appears to interact differently with FCAPs and UFCAPs, resulting in varied cardiac changes, which suggests that the cardiovascular effects of particle-gas co-exposures are not simply additive or even generalizable. Additionally, the mode of toxicity underlying this effect may be subtle given none of the exposures described here impaired post-ischemia recovery. Electronic supplementary material The online version of this article (doi:10.1186/s12989-014-0054-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nicole Kurhanewicz
- Curriculum in Toxicology, School of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - Rachel McIntosh-Kastrinsky
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - Haiyan Tong
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park, Chapel Hill, NC, 27711, USA.
| | - Leon Walsh
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park, Chapel Hill, NC, 27711, USA.
| | - Aimen K Farraj
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park, Chapel Hill, NC, 27711, USA.
| | - Mehdi S Hazari
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park, Chapel Hill, NC, 27711, USA.
| |
Collapse
|
26
|
Abstract
Cardiac regeneration strategies and de novo generation of cardiomyocytes have long been significant areas of research interest in cardiovascular medicine. In this review, we outline a variety of common cell sources and methods used to regenerate cardiomyocytes and highlight the important role that key Circulation Research articles have played in this flourishing field.
Collapse
Affiliation(s)
- Elena Matsa
- From the Stanford Cardiovascular Institute, Departments of Medicine and Radiology, Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | | | | |
Collapse
|
27
|
Li G, Cheng G, Wu J, Ma S, Sun C. New iPSC for old long QT syndrome modeling: putting the evidence into perspective. Exp Biol Med (Maywood) 2013; 239:131-40. [PMID: 24363251 DOI: 10.1177/1535370213514000] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Induced pluripotent stem cells (iPS cells or iPSCs) are typically derived by transfection of certain stem cell-associated genes into non-pluripotent cells, such as adult fibroblasts (typically adult somatic cells). Various diseases can be modeled through iPSC technology. The important implication of iPSCs to offer an unprecedented opportunity to recapitulate pathologic human tissue formation in vitro has generated great excitement and interest in the whole biomedical research community. Long QT syndrome (LQTS), an inherited heart disease, is characterized by prolonged QT interval on a surface electrocardiogram. LQTS presents with life-threatening cardiac arrhythmias, which can lead to fainting, syncope, and sudden death. The iPSC-derived cardiomyocytes from LQTS patients offer a potentially unlimited source of materials for biomedical study. They can be used to recapitulate complex physiological phenotypes, probe toxicological testing and drug screening, clarify the novel mechanistic insights and may also rectify gene defects at the cellular and molecular level. Despite the emerging challenges, iPSC technology has been increasingly recognized as a valuable and growing toolkit for modeling LQTS over other various models of human diseases.
Collapse
Affiliation(s)
- Guoliang Li
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | | | | | | | | |
Collapse
|
28
|
Kapur S, Macrae CA. The developmental basis of adult arrhythmia: atrial fibrillation as a paradigm. Front Physiol 2013; 4:221. [PMID: 24062689 PMCID: PMC3771314 DOI: 10.3389/fphys.2013.00221] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 08/02/2013] [Indexed: 11/17/2022] Open
Abstract
Normal cardiac rhythm is one of the most fundamental physiologic phenomena, emerging early in the establishment of the vertebrate body plan. The developmental pathways underlying the patterning and maintenance of stable cardiac electrophysiology must be extremely robust, but are only now beginning to be unraveled. The step-wise emergence of automaticity, AV delay and sequential conduction are each tightly regulated and perturbations of these patterning events is now known to play an integral role in pediatric and adult cardiac arrhythmias. Electrophysiologic patterning within individual cardiac chambers is subject to exquisite control and is influenced by early physiology superimposed on the underlying gene networks that regulate cardiogenesis. As additional cell populations migrate to the developing heart these too bring further complexity to the organ, as it adapts to the dynamic requirements of a growing organism. A comprehensive understanding of the developmental basis of normal rhythm will inform not only the mechanisms of inherited arrhythmias, but also the differential regional propensities of the adult heart to acquired arrhythmias. In this review we use atrial fibrillation as a generalizable example where the various factors are perhaps best understood.
Collapse
Affiliation(s)
- Sunil Kapur
- Medicine, Cardiovascular Division, Brigham and Women's Hospital and Harvard Medical School Boston, MA, USA
| | | |
Collapse
|
29
|
|
30
|
Affiliation(s)
- Calum A MacRae
- Harvard Medical School, Brigham and Women's Hospital, Cardiovascular Division,
75 Francis Street, Boston, MA 02115, P-857 307 0301, F-857 307 0300, USA
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW A substantial genetic contribution to the etiology of atrial fibrillation has emerged in the last decade, and has bolstered links between this arrhythmia and other forms of heart disease. In this article, we will summarize the work that has defined the inherited diathesis toward atrial fibrillation, outline the genetic studies to date, and characterize the roadblocks to a complete mechanistic understanding of this common arrhythmia. RECENT FINDINGS Clinical genetic work has demonstrated a large heritable contribution in atrial fibrillation, with studies in lone forms of the arrhythmia suggesting a traditional monogenic syndrome with reduced penetrance. Several Mendelian loci for typical forms of atrial fibrillation have been identified but the genes have not yet been cloned. Rare forms of familial atrial fibrillation are caused by mutations in potassium channel genes, and there are single families with mutations in a nuclear pore and a natriuretic peptide gene, respectively. Common loci with small effects are now being identified in genome-wide association (GWA) studies, including a locus on chromosome 4q25. These loci explain less than 10% of the inherited contribution to the arrhythmia, suggesting there are major contributions that have not yet been uncovered. SUMMARY Although great strides have been made in exploring the genetics of atrial fibrillation, the paroxysmal and asymptomatic nature of the phenotype challenges investigators as they seek the mechanistic basis of the arrhythmia.
Collapse
|
32
|
Niederer SA, Land S, Omholt SW, Smith NP. Interpreting genetic effects through models of cardiac electromechanics. Am J Physiol Heart Circ Physiol 2012; 303:H1294-303. [PMID: 23042948 DOI: 10.1152/ajpheart.00121.2012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Multiscale models of cardiac electromechanics are being increasingly focused on understanding how genetic variation and environment underpin multiple disease states. In this paper we review the current state of the art in both the development of specific models and the physiological insights they have produced. This growing research body includes the development of models for capturing the effects of changes in function in both single and multiple proteins in both specific expression systems and in vivo contexts. Finally, the potential for using this approach for ultimately predicting phenotypes from genetic sequence information is discussed.
Collapse
Affiliation(s)
- S A Niederer
- Department of Biomedical Engineering, King's College London, King's Health Partners, Saint Thomas' Hospital, London, UK
| | | | | | | |
Collapse
|
33
|
Blazeski A, Zhu R, Hunter DW, Weinberg SH, Zambidis ET, Tung L. Cardiomyocytes derived from human induced pluripotent stem cells as models for normal and diseased cardiac electrophysiology and contractility. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2012; 110:166-77. [PMID: 22971665 PMCID: PMC3910285 DOI: 10.1016/j.pbiomolbio.2012.07.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 07/30/2012] [Indexed: 12/21/2022]
Abstract
Since the first description of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), these cells have garnered tremendous interest for their potential use in patient-specific analysis and therapy. Additionally, hiPSC-CMs can be derived from donor cells from patients with specific cardiac disorders, enabling in vitro human disease models for mechanistic study and therapeutic drug assessment. However, a full understanding of their electrophysiological and contractile function is necessary before this potential can be realized. Here, we review this emerging field from a functional perspective, with particular emphasis on beating rate, action potential, ionic currents, multicellular conduction, calcium handling and contraction. We further review extant hiPSC-CM disease models that recapitulate genetic myocardial disease.
Collapse
Affiliation(s)
- Adriana Blazeski
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| | - Renjun Zhu
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| | - David W. Hunter
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| | - Seth H. Weinberg
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| | - Elias T. Zambidis
- Institute for Cell Engineering and Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University, Baltimore, MD
| | - Leslie Tung
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| |
Collapse
|
34
|
Blazeski A, Zhu R, Hunter DW, Weinberg SH, Boheler KR, Zambidis ET, Tung L. Electrophysiological and contractile function of cardiomyocytes derived from human embryonic stem cells. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2012; 110:178-95. [PMID: 22958937 DOI: 10.1016/j.pbiomolbio.2012.07.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 07/30/2012] [Indexed: 12/23/2022]
Abstract
Human embryonic stem cells have emerged as the prototypical source from which cardiomyocytes can be derived for use in drug discovery and cell therapy. However, such applications require that these cardiomyocytes (hESC-CMs) faithfully recapitulate the physiology of adult cells, especially in relation to their electrophysiological and contractile function. We review what is known about the electrophysiology of hESC-CMs in terms of beating rate, action potential characteristics, ionic currents, and cellular coupling as well as their contractility in terms of calcium cycling and contraction. We also discuss the heterogeneity in cellular phenotypes that arises from variability in cardiac differentiation, maturation, and culture conditions, and summarize present strategies that have been implemented to reduce this heterogeneity. Finally, we present original electrophysiological data from optical maps of hESC-CM clusters.
Collapse
Affiliation(s)
- Adriana Blazeski
- Department of Biomedical Engineering, The Johns Hopkins University, 720 Rutland Ave., Baltimore, MD 21205, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Yu F, Zhao Y, Gu J, Quigley KL, Chi NC, Tai YC, Hsiai TK. Flexible microelectrode arrays to interface epicardial electrical signals with intracardial calcium transients in zebrafish hearts. Biomed Microdevices 2012; 14:357-66. [PMID: 22124886 DOI: 10.1007/s10544-011-9612-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The zebrafish (Danio rerio) is an emerging genetic model for regenerative medicine. In humans, myocardial infarction results in the irreversible loss of cardiomyocytes. However, zebrafish hearts fully regenerate after a 20% ventricular resection, without either scarring or arrhythmias. To study this cardiac regeneration, we developed implantable flexible multi-microelectrode membrane arrays that measure the epicardial electrocardiogram signals of zebrafish in real-time. The microelectrode electrical signals allowed for a high level of both temporal and spatial resolution (~20 μm), and the signal to noise ratio of the epicardial ECG was comparable to that of surface electrode ECG (7.1 dB vs. 7.4 dB, respectively). Processing and analysis of the signals from the microelectrode array demonstrated distinct ECG signals: namely, atrial conduction (P waves), ventricular contraction (QRS), and ventricular repolarization (QT interval). The electrical signals were in synchrony with optically measured Calcium concentration gradients in terms of d[Ca²⁺]/dt at both whole heart and tissue levels. These microelectrodes therefore provide a real-time analytical tool for monitoring conduction phenotypes of small vertebral animals with a high temporal and spatial resolution.
Collapse
Affiliation(s)
- Fei Yu
- Biomedical Engineering and Cardiovascular Medicine, University of Southern California, Los Angeles, CA, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Affiliation(s)
- Andrew J Sauer
- Center for Human Genetic Research, Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
| | | |
Collapse
|
37
|
Abstract
As the current paradigms of drug discovery evolve, it has become clear that a more comprehensive understanding of the interactions between small molecules and organismal biology will be vital. The zebrafish is emerging as a complement to existing in vitro technologies and established preclinical in vivo models that can be scaled for high-throughput. In this review, we highlight the current status of zebrafish toxicology studies, identify potential future niches for the model in the drug development pipeline, and define the hurdles that must be overcome as zebrafish technologies are refined for systematic toxicology.
Collapse
Affiliation(s)
- Randall T Peterson
- Harvard Medical School, Massachusetts General Hospital, and Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
38
|
Barbuti A. Virus-free iPS-derived cardiomyocytes: a new piece in the puzzle of patient-tailored therapies. Cardiovasc Res 2011; 91:559-60. [PMID: 21742676 DOI: 10.1093/cvr/cvr199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
39
|
Yu F, Huang J, Adlerz K, Jadvar H, Hamdan MH, Chi N, Chen JN, Hsiai TK. Evolving cardiac conduction phenotypes in developing zebrafish larvae: implications to drug sensitivity. Zebrafish 2010; 7:325-31. [PMID: 20958244 DOI: 10.1089/zeb.2010.0658] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cardiac arrhythmias include problems with impulse formation and/or conduction abnormalities. Zebrafish (Danio rerio) is an emerging model system for studying the cardiac conduction system. However, real-time recording of the electrocardiogram remains a challenge. In the present study, we assessed the feasibility of recording electrical cardiogram (ECG) signals from the zebrafish larvae using the micropipette electrodes, and demonstrated the dynamic changes in ECG signals and their sensitivity to Amiodarone during the developmental stages. We observed that ECG signals revealed P waves and QRS complexes at 7 days postfertilization (dpf). T waves started to develop at 14 dpf. Distinct P waves, QRS complexes, and T waves were similar to those of adult zebrafish at 35 dpf, accompanied by a statistically significant decrease in QRS intervals (from 256 ± 16 ms at 7 dpf to 54 ± 6 ms, p < 0.01, n = 5). In response to Amiodarone, ECG signals showed QRS prolongation from 7 to 35 dpf (p < 0.05, n = 5). Hence, micropipette electrodes can be applied to detect evolving ECG signals from the developing zebrafish larvae, thus providing a noninvasive and nonparalyzing approach to investigate cardiac conduction phenotypes in response to genetic, epigenetic, or pharmacologic perturbation.
Collapse
Affiliation(s)
- Fei Yu
- Department of Biomedical Engineering and Cardiovascular Medicine, University of Southern California, Los Angeles, California 90089, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) originally emerged as a pathologic diagnosis based on distinctive autopsy findings in cases of premature sudden death. Subsequently these characteristic pathologic features were associated with ventricular tachycardia of right ventricular origin and syncope. ARVC is a rare condition and our understanding of the disorder has been confounded by multiple small, highly selected series. Driven by both family studies and improved non-invasive imaging tools the clinical diagnosis of ARVC has broadened, in some instances extending far beyond the original limits of the syndrome. In recent years false-positive diagnoses have increased, thus stimulating investigators to move toward more rigorous clinical criteria. Despite the efforts of a Task Force to establish a baseline for subsequent empiric testing, these criteria have often inadvertently been used as a definitive diagnostic tool in the absence of prospective data. Recent genetic studies have revealed substantial etiologic heterogeneity, and ARVC is emerging as a syndrome consisting of multiple discrete disease entities, in part explaining the tremendous variation in clinical features and natural history seen in prior reports.
Collapse
Affiliation(s)
- Patrick T Ellinor
- Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital, Charlestown, Boston, MA, USA
| | | | | |
Collapse
|
41
|
Macrae CA. Cardiac Arrhythmia: In vivo screening in the zebrafish to overcome complexity in drug discovery. Expert Opin Drug Discov 2010; 5:619-632. [PMID: 20835353 DOI: 10.1517/17460441.2010.492826] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
IMPORTANCE OF THE FIELD: Cardiac arrhythmias remain a major challenge for modern drug discovery. Clinical events are paroxysmal, often rare and may be asymptomatic until a highly morbid complication. Target selection is often based on limited information and though highly specific agents are identified in screening, the final efficacy is often compromised by unanticipated systemic responses, a narrow therapeutic index and substantial toxicities. AREAS COVERED IN THIS REVIEW: Our understanding of complexity of arrhythmogenesis has grown dramatically over the last two decades, and the range of potential disease mechanisms now includes pathways previously thought only tangentially involved in arrhythmia. This review surveys the literature on arrhythmia mechanisms from 1965 to the present day, outlines the complex biology underlying potentially each and every rhythm disturbance, and highlights the problems for rational target identification. The rationale for in vivo screening is described and the utility of the zebrafish for this approach and for complementary work in functional genomics is discussed. Current limitations of the model in this setting and the need for careful validation in new disease areas are also described. WHAT THE READER WILL GAIN: An overview of the complex mechanisms underlying most clinical arrhythmias, and insight into the limits of ion channel conductances as drug targets. An introduction to the zebrafish as a model organism, in particular for cardiovascular biology. Potential approaches to overcoming the hurdles to drug discovery in the face of complex biology including in vivo screening of zebrafish genetic disease models. TAKE HOME MESSAGE: In vivo screening in faithful disease models allows the effects of drugs on integrative physiology and disease biology to be captured during the screening process, in a manner agnostic to potential drug target or targets. This systematic strategy bypasses current gaps in our understanding of disease biology, but emphasizes the importance of the rigor of the disease model.
Collapse
Affiliation(s)
- Calum A Macrae
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, The Broad Institute of MIT and Harvard
| |
Collapse
|
42
|
Yu F, Li R, Parks E, Takabe W, Hsiai TK. Electrocardiogram signals to assess zebrafish heart regeneration: implication of long QT intervals. Ann Biomed Eng 2010; 38:2346-57. [PMID: 20221900 DOI: 10.1007/s10439-010-9993-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Accepted: 03/02/2010] [Indexed: 12/01/2022]
Abstract
Zebrafish is an emerging model system for cardiac conduction and regeneration. Zebrafish heart regenerates after 20% ventricular resection within 60 days. Whether cardiac conduction phenotype correlated with cardiomyocyte regeneration remained undefined. Longitudinal monitoring of the adult zebrafish heart (n = 12) was performed in terms of atrial contraction (PR intervals), ventricular depolarization (QRS complex) and repolarization (heart rated corrected QTc interval). Baseline electrocardiogram (ECG) signals were recorded one day prior to resection and twice per week over 59 days. Immunostaining for gap junctions with anti-Connexin-43 antibody was compared between the sham (n = 5) and ventricular resection at 60 days post-resection (dpr) (n = 7). Heart rate variability, QTc prolongation and J-point depression developed in the resected group but not in the sham. Despite a trend toward heart rate variability in response to ventricular resection, the differences between the resected and sham fish were, by and large, statistically insignificant. At 10 dpr, J-point depression was statistically significant (sham: -0.179 +/- 0.061 mV vs. ventricular resection: -0.353 +/- 0.105 mV, p < 0.01, n = 7). At 60 days, histology revealed either cardiomyocyte regeneration (n = 4) or scar tissues (n = 3). J-point depression was no longer statistically significant at 59 dpr (sham: -0.114 +/- 0.085 mV; scar tissue: -0.268 +/- 0.178 mV, p > 0.05, n = 3; regeneration: -0.209 +/- 0.119 mV, p > 0.05, n = 4). Despite positive Connexin-43 staining in the regeneration group, QTc intervals remained prolonged (sham: 325 +/- 42 ms, n = 5; scar tissues: 534 +/- 51 ms, p < 0.01, n = 3; regeneration: 496 +/- 31 ms, p < 0.01, n = 4). Thus, we observed delayed electric repolarization in either the regenerated hearts or scar tissues. Moreover, early regenerated cardiomyocytes lacked the conduction phenotypes of the sham fish.
Collapse
Affiliation(s)
- Fei Yu
- Department of Biomedical Engineering and Division of Cardiovascular Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | | | | | | | | |
Collapse
|
43
|
Scholz EP, Niemer N, Hassel D, Zitron E, Bürgers HF, Bloehs R, Seyler C, Scherer D, Thomas D, Kathöfer S, Katus HA, Rottbauer WA, Karle CA. Biophysical properties of zebrafish ether-à-go-go related gene potassium channels. Biochem Biophys Res Commun 2009; 381:159-64. [DOI: 10.1016/j.bbrc.2009.02.042] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Accepted: 02/02/2009] [Indexed: 11/29/2022]
|
44
|
Micro-electrocardiograms to study post-ventricular amputation of zebrafish heart. Ann Biomed Eng 2009; 37:890-901. [PMID: 19280341 DOI: 10.1007/s10439-009-9668-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Accepted: 03/03/2009] [Indexed: 12/11/2022]
Abstract
The zebrafish (Danio rerio) is an emerging model for cardiovascular research. The zebrafish heart regenerates after 20% ventricular amputation. However, assessment of the physiological responses during heart regeneration has been hampered by the small size of the heart and the necessity of conducting experiments in an aqueous environment. We developed a methodology to monitor a real-time surface electrocardiogram (ECG) by the use of micro-electrodes, signal amplification, and a low pass-filter at a sampling rate of 1 kHz. Wavelet transform was used to further remove ambient noises. Rather than paralyzing the fish, we performed mild sedation by placing the fish in a water bath mixed with MS-222 (tricane methanesulfonate). We recorded distinct P waves for atrial contraction, QRS complexes for ventricular depolarization, and QT intervals for ventricular repolarization prior to, and 2 and 4 days post-amputation (dpa). Sedation reduced the mean fish heart rate from 149 +/- 18 to 90 +/- 17 beats/min. The PR and QRS intervals remained unchanged in response to ventricular apical amputation (n = 6, p > 0.05). Corrected QT intervals (QTc) were shortened 4 dpa (n = 6, p < 0.05). In a parallel study, histology revealed that apical thrombi were replaced with fibrin clots and collagen fibers. Atrial arrhythmia was noted in response to prolonged sedation. Unlike the human counterpart, ventricular tachycardia or fibrillation was not observed in response to ventricular amputation 2 and 4 dpa. Taken together, we demonstrated a minimally invasive methodology to monitor zebrafish heart function, electrical activities, and regeneration in real-time.
Collapse
|
45
|
Ellinor PT, Choudry S, Macrae CA. Genetics of atrial fibrillation. Future Cardiol 2006; 2:579-84. [PMID: 19804195 DOI: 10.2217/14796678.2.5.579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Atrial fibrillation (AF) is the most common arrhythmia encountered in clinical practice. Despite its prevalence, relatively little is known regarding the primary mechanisms of AF and, therefore, current treatment practices focus mainly on controlling the disorder and preventing its complications once it is already present. The study of the pathogenesis of AF is complicated by the varied clinical presentation of the arrhythmia and its coexistence with other cardiac pathologies. This article reviews current efforts to delineate the fundamental mechanisms of AF, with a focus on genetic studies. Identification of the underlying etiology may result in the development of more targeted and effective therapies for AF.
Collapse
Affiliation(s)
- Patrick T Ellinor
- Massachusetts General Hospital, Cardiac Arrhythmia Service & Cardiovascular Research Center, 55 Fruit Street, GRB 109, Boston, MA 02114, USA.
| | | | | |
Collapse
|