1
|
Pedroni A, Yilmaz E, Del Vecchio L, Bhattarai P, Vidal IT, Dai YWE, Koutsogiannis K, Kizil C, Ampatzis K. Decoding the molecular, cellular, and functional heterogeneity of zebrafish intracardiac nervous system. Nat Commun 2024; 15:10483. [PMID: 39632839 PMCID: PMC11618350 DOI: 10.1038/s41467-024-54830-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024] Open
Abstract
The proper functioning of the heart relies on the intricate interplay between the central nervous system and the local neuronal networks within the heart itself. While the central innervation of the heart has been extensively studied, the organization and functionality of the intracardiac nervous system (IcNS) remain largely unexplored. Here, we present a comprehensive taxonomy of the IcNS, utilizing single-cell RNA sequencing, anatomical studies, and electrophysiological techniques. Our findings reveal a diverse array of neuronal types within the IcNS, exceeding previous expectations. We identify a subset of neurons exhibiting characteristics akin to pacemaker/rhythmogenic neurons similar to those found in Central Pattern Generator networks of the central nervous system. Our results underscore the heterogeneity within the IcNS and its key role in regulating the heart's rhythmic functionality. The classification and characterization of the IcNS presented here serve as a valuable resource for further exploration into the mechanisms underlying heart functionality and the pathophysiology of associated cardiac disorders.
Collapse
Affiliation(s)
- Andrea Pedroni
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Elanur Yilmaz
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
- Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
| | - Lisa Del Vecchio
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Prabesh Bhattarai
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
- Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
| | - Inés Talaya Vidal
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Yu-Wen E Dai
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | | | - Caghan Kizil
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA.
- Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA.
| | | |
Collapse
|
2
|
Abu Nahia K, Sulej A, Migdał M, Ochocka N, Ho R, Kamińska B, Zagorski M, Winata CL. scRNA-seq reveals the diversity of the developing cardiac cell lineage and molecular players in heart rhythm regulation. iScience 2024; 27:110083. [PMID: 38872974 PMCID: PMC11170199 DOI: 10.1016/j.isci.2024.110083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/26/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Abstract
We utilized scRNA-seq to delineate the diversity of cell types in the zebrafish heart. Transcriptome profiling of over 50,000 cells at 48 and 72 hpf defined at least 18 discrete cell lineages of the developing heart. Utilizing well-established gene signatures, we identified a population of cells likely to be the primary pacemaker and characterized the transcriptome profile defining this critical cell type. Two previously uncharacterized genes, atp1b3b and colec10, were found to be enriched in the sinoatrial cardiomyocytes. CRISPR/Cas9-mediated knockout of these two genes significantly reduced heart rate, implicating their role in cardiac development and conduction. Additionally, we describe other cardiac cell lineages, including the endothelial and neural cells, providing their expression profiles as a resource. Our results established a detailed atlas of the developing heart, providing valuable insights into cellular and molecular mechanisms, and pinpointed potential new players in heart rhythm regulation.
Collapse
Affiliation(s)
- Karim Abu Nahia
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Agata Sulej
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Maciej Migdał
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Natalia Ochocka
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Richard Ho
- Institute of Theoretical Physics and Mark Kac Center for Complex Systems Research, Jagiellonian University, Cracow, Poland
- The Njord Centre, Department of Physics, University of Oslo, Oslo, Norway
| | - Bożena Kamińska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Marcin Zagorski
- Institute of Theoretical Physics and Mark Kac Center for Complex Systems Research, Jagiellonian University, Cracow, Poland
| | | |
Collapse
|
3
|
Stoyek MR, Doane SE, Dallaire SE, Long ZD, Ramia JM, Cassidy-Nolan DL, Poon KL, Brand T, Quinn TA. POPDC1 Variants Cause Atrioventricular Node Dysfunction and Arrhythmogenic Changes in Cardiac Electrophysiology and Intracellular Calcium Handling in Zebrafish. Genes (Basel) 2024; 15:280. [PMID: 38540339 PMCID: PMC10969970 DOI: 10.3390/genes15030280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 06/15/2024] Open
Abstract
Popeye domain-containing (POPDC) proteins selectively bind cAMP and mediate cellular responses to sympathetic nervous system (SNS) stimulation. The first discovered human genetic variant (POPDC1S201F) is associated with atrioventricular (AV) block, which is exacerbated by increased SNS activity. Zebrafish carrying the homologous mutation (popdc1S191F) display a similar phenotype to humans. To investigate the impact of POPDC1 dysfunction on cardiac electrophysiology and intracellular calcium handling, homozygous popdc1S191F and popdc1 knock-out (popdc1KO) zebrafish larvae and adult isolated popdc1S191F hearts were studied by functional fluorescent analysis. It was found that in popdc1S191F and popdc1KO larvae, heart rate (HR), AV delay, action potential (AP) and calcium transient (CaT) upstroke speed, and AP duration were less than in wild-type larvae, whereas CaT duration was greater. SNS stress by β-adrenergic receptor stimulation with isoproterenol increased HR, lengthened AV delay, slowed AP and CaT upstroke speed, and shortened AP and CaT duration, yet did not result in arrhythmias. In adult popdc1S191F zebrafish hearts, there was a higher incidence of AV block, slower AP upstroke speed, and longer AP duration compared to wild-type hearts, with no differences in CaT. SNS stress increased AV delay and led to further AV block in popdc1S191F hearts while decreasing AP and CaT duration. Overall, we have revealed that arrhythmogenic effects of POPDC1 dysfunction on cardiac electrophysiology and intracellular calcium handling in zebrafish are varied, but already present in early development, and that AV node dysfunction may underlie SNS-induced arrhythmogenesis associated with popdc1 mutation in adults.
Collapse
Affiliation(s)
- Matthew R. Stoyek
- Department of Physiology & Biophysics, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.S.); (S.E.D.); (S.E.D.); (Z.D.L.); (J.M.R.); (D.L.C.-N.)
| | - Sarah E. Doane
- Department of Physiology & Biophysics, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.S.); (S.E.D.); (S.E.D.); (Z.D.L.); (J.M.R.); (D.L.C.-N.)
| | - Shannon E. Dallaire
- Department of Physiology & Biophysics, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.S.); (S.E.D.); (S.E.D.); (Z.D.L.); (J.M.R.); (D.L.C.-N.)
| | - Zachary D. Long
- Department of Physiology & Biophysics, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.S.); (S.E.D.); (S.E.D.); (Z.D.L.); (J.M.R.); (D.L.C.-N.)
| | - Jessica M. Ramia
- Department of Physiology & Biophysics, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.S.); (S.E.D.); (S.E.D.); (Z.D.L.); (J.M.R.); (D.L.C.-N.)
| | - Donovan L. Cassidy-Nolan
- Department of Physiology & Biophysics, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.S.); (S.E.D.); (S.E.D.); (Z.D.L.); (J.M.R.); (D.L.C.-N.)
| | - Kar-Lai Poon
- National Heart & Lung Institute, Imperial College London, London W12 0NN, UK; (K.-L.P.); (T.B.)
| | - Thomas Brand
- National Heart & Lung Institute, Imperial College London, London W12 0NN, UK; (K.-L.P.); (T.B.)
| | - T. Alexander Quinn
- Department of Physiology & Biophysics, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.S.); (S.E.D.); (S.E.D.); (Z.D.L.); (J.M.R.); (D.L.C.-N.)
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
4
|
Da Silveira Cavalcante L, Higuita ML, González-Rosa JM, Marques B, To S, Pendexter CA, Cronin SE, Gopinathan K, de Vries RJ, Ellett F, Uygun K, Langenau DM, Toner M, Tessier SN. Zebrafish as a high throughput model for organ preservation and transplantation research. FASEB J 2023; 37:e23187. [PMID: 37718489 PMCID: PMC10754348 DOI: 10.1096/fj.202300076r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 08/15/2023] [Accepted: 08/24/2023] [Indexed: 09/19/2023]
Abstract
Despite decades of effort, the preservation of complex organs for transplantation remains a significant barrier that exacerbates the organ shortage crisis. Progress in organ preservation research is significantly hindered by suboptimal research tools that force investigators to sacrifice translatability over throughput. For instance, simple model systems, such as single cell monolayers or co-cultures, lack native tissue structure and functional assessment, while mammalian whole organs are complex systems with confounding variables not compatible with high-throughput experimentation. In response, diverse fields and industries have bridged this experimental gap through the development of rich and robust resources for the use of zebrafish as a model organism. Through this study, we aim to demonstrate the value zebrafish pose for the fields of solid organ preservation and transplantation, especially with respect to experimental transplantation efforts. A wide array of methods were customized and validated for preservation-specific experimentation utilizing zebrafish, including the development of assays at multiple developmental stages (larvae and adult), methods for loading and unloading preservation agents, and the development of viability scores to quantify functional outcomes. Using this platform, the largest and most comprehensive screen of cryoprotectant agents (CPAs) was performed to determine their toxicity and efficiency at preserving complex organ systems using a high subzero approach called partial freezing (i.e., storage in the frozen state at -10°C). As a result, adult zebrafish cardiac function was successfully preserved after 5 days of partial freezing storage. In combination, the methods and techniques developed have the potential to drive and accelerate research in the fields of solid organ preservation and transplantation.
Collapse
Affiliation(s)
- Luciana Da Silveira Cavalcante
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| | - Manuela Lopera Higuita
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| | - Juan Manuel González-Rosa
- Cardiovascular Research Center, Massachusetts General Hospital Research Institute, Harvard Medical School, Charlestown MA, USA
| | - Beatriz Marques
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
| | - Samantha To
- Cardiovascular Research Center, Massachusetts General Hospital Research Institute, Harvard Medical School, Charlestown MA, USA
| | - Casie A. Pendexter
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| | - Stephanie E.J. Cronin
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| | - Kaustav Gopinathan
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
| | - Reinier J. de Vries
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| | - Felix Ellett
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| | - Korkut Uygun
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| | - David M. Langenau
- Molecular Pathology Unit and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Mehmet Toner
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| | - Shannon N. Tessier
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Boston MA, USA
- Shriners Hospitals for Children - Boston, Boston MA, USA
| |
Collapse
|
5
|
Baillie JS, Gendernalik A, Garrity DM, Bark D, Quinn TA. The in vivo study of cardiac mechano-electric and mechano-mechanical coupling during heart development in zebrafish. Front Physiol 2023; 14:1086050. [PMID: 37007999 PMCID: PMC10060984 DOI: 10.3389/fphys.2023.1086050] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
In the adult heart, acute adaptation of electrical and mechanical activity to changes in mechanical load occurs via feedback processes known as “mechano-electric coupling” and “mechano-mechanical coupling.” Whether this occurs during cardiac development is ill-defined, as acutely altering the heart’s mechanical load while measuring functional responses in traditional experimental models is difficult, as embryogenesis occurs in utero, making the heart inaccessible. These limitations can be overcome with zebrafish, as larvae develop in a dish and are nearly transparent, allowing for in vivo manipulation and measurement of cardiac structure and function. Here we present a novel approach for the in vivo study of mechano-electric and mechano-mechanical coupling in the developing zebrafish heart. This innovative methodology involves acute in vivo atrial dilation (i.e., increased atrial preload) in larval zebrafish by injection of a controlled volume into the venous circulation immediately upstream of the heart, combined with optical measurement of the acute electrical (change in heart rate) and mechanical (change in stroke area) response. In proof-of-concept experiments, we applied our new method to 48 h post-fertilisation zebrafish, which revealed differences between the electrical and mechanical response to atrial dilation. In response to an acute increase in atrial preload there is a large increase in atrial stroke area but no change in heart rate, demonstrating that in contrast to the fully developed heart, during early cardiac development mechano-mechanical coupling alone drives the adaptive increase in atrial output. Overall, in this methodological paper we present our new experimental approach for the study of mechano-electric and mechano-mechanical coupling during cardiac development and demonstrate its potential for understanding the essential adaptation of heart function to acute changes in mechanical load.
Collapse
Affiliation(s)
| | - Alex Gendernalik
- Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
| | | | - David Bark
- Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
- Mechanical Engineering, Colorado State University, Fort Collins, CO, United States
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
| | - T. Alexander Quinn
- Physiology & Biophysics, Dalhousie University, Halifax, NS, Canada
- Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
- *Correspondence: T. Alexander Quinn,
| |
Collapse
|
6
|
Welzel G, Schuster S. Electric catfish hearts are not intrinsically immune to electric shocks. J Exp Biol 2022; 225:276258. [PMID: 35946177 DOI: 10.1242/jeb.244307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/19/2022] [Indexed: 10/15/2022]
Abstract
High voltage electric shocks cause life threatening cardiac injuries such as sudden cardiac standstill or severe myocardial injury. Here, we analysed the physiology of the heart of the strongly electric catfish (Malapterurus beninensis) that stuns prey with high-voltage shocks but is immune to its own, as well as external, high-voltage shocks. Neither a detailed analysis of the electrocardiogram nor the structure of the heart indicated a specialized cardiac conduction system. Using a suitable perfusion system, we discovered that, despite its immunity in vivo, the explanted heart of electric catfish can readily be activated by external electrical currents and is equally sensitive to electric shock-induced arrhythmias as similar-sized goldfish hearts. The surprise thus is that the electric catfish has a vulnerable heart that requires to be protected by highly efficient but presently unknown means.
Collapse
Affiliation(s)
- Georg Welzel
- Department of Animal Physiology, University of Bayreuth, Universitätsstrasse 30, 95447 Bayreuth, Germany
| | - Stefan Schuster
- Department of Animal Physiology, University of Bayreuth, Universitätsstrasse 30, 95447 Bayreuth, Germany
| |
Collapse
|
7
|
Joyce W, Pan YK, Garvey K, Saxena V, Perry S. Regulation of heart rate following genetic deletion of the ß1 adrenergic receptor in larval zebrafish. Acta Physiol (Oxf) 2022; 235:e13849. [PMID: 35665450 PMCID: PMC9539580 DOI: 10.1111/apha.13849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/17/2022] [Accepted: 06/01/2022] [Indexed: 12/01/2022]
Abstract
Aim Although zebrafish are gaining popularity as biomedical models of cardiovascular disease, our understanding of their cardiac control mechanisms is fragmentary. Our goal was to clarify the controversial role of the ß1‐adrenergic receptor (AR) in the regulation of heart rate in zebrafish. Methods CRISPR‐Cas9 was used to delete the adrb1 gene in zebrafish allowing us to generate a stable adrb1−/− line. Larval heart rates were measured during pharmacological protocols and with exposure to hypercapnia. Expression of the five zebrafish adrb genes were measured in larval zebrafish hearts using qPCR. Results Compared with genetically matched wild‐types (adrb1+/+), adrb1−/− larvae exhibited ~20 beats min−1 lower heart rate, measured from 2 to 21 days post‐fertilization (dpf). Nevertheless, adrb1−/− larvae exhibited preserved positive chronotropic responses to pharmacological treatment with AR agonists (adrenaline, noradrenaline, isoproterenol), which were blocked by propranolol (general ß‐AR antagonist). Regardless of genotype, larvae exhibited similar increases in heart rate in response to hypercapnia (1% CO2) at 5 dpf, but tachycardia was blunted in adrb1−/− larvae at 6 dpf. adrb1 gene expression was abolished in the hearts of adrb1−/− larvae, confirming successful knockout. While gene expression of adrb2a and adrb3a was unchanged, adrb2b and adrb3b mRNA levels increased in adrb1−/− larval hearts. Conclusion Despite adrb1 contributing to the setting of resting heart rate in larvae, it is not strictly essential for zebrafish, as we generated a viable and breeding adrb1−/− line. The chronotropic effects of adrenergic stimulation persist in adrb1−/− zebrafish, likely due to the upregulation of other ß‐AR subtypes.
Collapse
Affiliation(s)
- William Joyce
- Department of Biology University of Ottawa Ottawa Ontario Canada
- Department of Biology – Zoophysiology Aarhus Universitet Aarhus C Denmark
| | - Yihang K. Pan
- Department of Biology University of Ottawa Ottawa Ontario Canada
| | - Kayla Garvey
- Department of Biology University of Ottawa Ottawa Ontario Canada
| | - Vishal Saxena
- Department of Biology University of Ottawa Ottawa Ontario Canada
| | - Steve F. Perry
- Department of Biology University of Ottawa Ottawa Ontario Canada
| |
Collapse
|
8
|
Arel E, Rolland L, Thireau J, Torrente AG, Bechard E, Bride J, Jopling C, Demion M, Le Guennec JY. The Effect of Hypothermia and Osmotic Shock on the Electrocardiogram of Adult Zebrafish. BIOLOGY 2022; 11:biology11040603. [PMID: 35453802 PMCID: PMC9025276 DOI: 10.3390/biology11040603] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/08/2022] [Accepted: 04/13/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary Assessing cardiac toxicity of new drugs is a requirement for their approval. One of the parameters which is carefully looked at is the QT interval, which is determined using an electrocardiogram (ECG). Before undertaking clinical trials using human patients, it is important to first perform pre-clinical tests using animal models. Zebrafish are widely used to study cardiac physiology and several reports suggest that although ECG measurement can be performed, the recording configuration appears to affect the results. Our research aimed to provide a comprehensive characterization of adult zebrafish ECG to determine the best practice for using this model during cardiac toxicity trials. We tested three recording configurations and determined that exposing the heart provided the most reliable and reproducible ECG recordings. We also determined the most accurate correction to apply to calculate the corrected QT, which makes the QT interval independent of the heart rate, a critical parameter when assessing drug cardiac toxicity. Overall, our study highlights the best conditions to record zebrafish ECG and demonstrates their utility for cardiac toxicity testing. Abstract The use of zebrafish to explore cardiac physiology has been widely adopted within the scientific community. Whether this animal model can be used to determine drug cardiac toxicity via electrocardiogram (ECG) analysis is still an ongoing question. Several reports indicate that the recording configuration severely affects the ECG waveforms and its derived-parameters, emphasizing the need for improved characterization. To address this problem, we recorded ECGs from adult zebrafish hearts in three different configurations (unexposed heart, exposed heart, and extracted heart) to identify the most reliable method to explore ECG recordings at baseline and in response to commonly used clinical therapies. We found that the exposed heart configuration provided the most reliable and reproducible ECG recordings of waveforms and intervals. We were unable to determine T wave morphology in unexposed hearts. In extracted hearts, ECG intervals were lengthened and P waves were unstable. However, in the exposed heart configuration, we were able to reliably record ECGs and subsequently establish the QT-RR relationship (Holzgrefe correction) in response to changes in heart rate.
Collapse
Affiliation(s)
- Elodie Arel
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, 34090 Montpellier, France; (E.A.); (J.T.); (E.B.); (J.B.); (M.D.); (J.-Y.L.G.)
- Institut de génomique fonctionnelle (IGF), Université de Montpellier, Inserm U1191, UMR CNRS 5203, 34094 Montpellier, France; (L.R.); (A.G.T.)
| | - Laura Rolland
- Institut de génomique fonctionnelle (IGF), Université de Montpellier, Inserm U1191, UMR CNRS 5203, 34094 Montpellier, France; (L.R.); (A.G.T.)
| | - Jérôme Thireau
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, 34090 Montpellier, France; (E.A.); (J.T.); (E.B.); (J.B.); (M.D.); (J.-Y.L.G.)
| | - Angelo Giovanni Torrente
- Institut de génomique fonctionnelle (IGF), Université de Montpellier, Inserm U1191, UMR CNRS 5203, 34094 Montpellier, France; (L.R.); (A.G.T.)
| | - Emilie Bechard
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, 34090 Montpellier, France; (E.A.); (J.T.); (E.B.); (J.B.); (M.D.); (J.-Y.L.G.)
| | - Jamie Bride
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, 34090 Montpellier, France; (E.A.); (J.T.); (E.B.); (J.B.); (M.D.); (J.-Y.L.G.)
- Institut de génomique fonctionnelle (IGF), Université de Montpellier, Inserm U1191, UMR CNRS 5203, 34094 Montpellier, France; (L.R.); (A.G.T.)
| | - Chris Jopling
- Institut de génomique fonctionnelle (IGF), Université de Montpellier, Inserm U1191, UMR CNRS 5203, 34094 Montpellier, France; (L.R.); (A.G.T.)
- Correspondence:
| | - Marie Demion
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, 34090 Montpellier, France; (E.A.); (J.T.); (E.B.); (J.B.); (M.D.); (J.-Y.L.G.)
| | - Jean-Yves Le Guennec
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, 34090 Montpellier, France; (E.A.); (J.T.); (E.B.); (J.B.); (M.D.); (J.-Y.L.G.)
| |
Collapse
|
9
|
Sacconi L, Silvestri L, Rodríguez EC, Armstrong GA, Pavone FS, Shrier A, Bub G. KHz-rate volumetric voltage imaging of the whole Zebrafish heart. BIOPHYSICAL REPORTS 2022; 2:100046. [PMID: 36425080 PMCID: PMC9680780 DOI: 10.1016/j.bpr.2022.100046] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/31/2022] [Indexed: 05/11/2023]
Abstract
Fast volumetric imaging is essential for understanding the function of excitable tissues such as those found in the brain and heart. Measuring cardiac voltage transients in tissue volumes is challenging, especially at the high spatial and temporal resolutions needed to give insight to cardiac function. We introduce a new imaging modality based on simultaneous illumination of multiple planes in the tissue and parallel detection with multiple cameras, avoiding compromises inherent in any scanning approach. The system enables imaging of voltage transients in situ, allowing us, for the first time to our knowledge, to map voltage activity in the whole heart volume at KHz rates. The high spatiotemporal resolution of our method enabled the observation of novel dynamics of electrical propagation through the zebrafish atrioventricular canal.
Collapse
Affiliation(s)
- Leonardo Sacconi
- European Laboratory for Non-linear Spectroscopy, and National Institute of Optics, National Research Council, Sesto Fiorentino, Italy
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Medical Faculty, University of Freiburg, Freiburg, Germany
- Corresponding author
| | - Ludovico Silvestri
- European Laboratory for Non-linear Spectroscopy, and National Institute of Optics, National Research Council, Sesto Fiorentino, Italy
- Department of Physics and Astronomy, University of Florence, Florence, Italy
| | | | - Gary A.B. Armstrong
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| | - Francesco S. Pavone
- European Laboratory for Non-linear Spectroscopy, and National Institute of Optics, National Research Council, Sesto Fiorentino, Italy
- Department of Physics and Astronomy, University of Florence, Florence, Italy
| | - Alvin Shrier
- Department of Physiology, McGill University, Montreal, Canada
| | - Gil Bub
- Department of Physiology, McGill University, Montreal, Canada
- Corresponding author
| |
Collapse
|
10
|
Stoyek MR, MacDonald EA, Mantifel M, Baillie JS, Selig BM, Croll RP, Smith FM, Quinn TA. Drivers of Sinoatrial Node Automaticity in Zebrafish: Comparison With Mechanisms of Mammalian Pacemaker Function. Front Physiol 2022; 13:818122. [PMID: 35295582 PMCID: PMC8919049 DOI: 10.3389/fphys.2022.818122] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/21/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiac excitation originates in the sinoatrial node (SAN), due to the automaticity of this distinct region of the heart. SAN automaticity is the result of a gradual depolarisation of the membrane potential in diastole, driven by a coupled system of transarcolemmal ion currents and intracellular Ca2+ cycling. The frequency of SAN excitation determines heart rate and is under the control of extra- and intracardiac (extrinsic and intrinsic) factors, including neural inputs and responses to tissue stretch. While the structure, function, and control of the SAN have been extensively studied in mammals, and some critical aspects have been shown to be similar in zebrafish, the specific drivers of zebrafish SAN automaticity and the response of its excitation to vagal nerve stimulation and mechanical preload remain incompletely understood. As the zebrafish represents an important alternative experimental model for the study of cardiac (patho-) physiology, we sought to determine its drivers of SAN automaticity and the response to nerve stimulation and baseline stretch. Using a pharmacological approach mirroring classic mammalian experiments, along with electrical stimulation of intact cardiac vagal nerves and the application of mechanical preload to the SAN, we demonstrate that the principal components of the coupled membrane- Ca2+ pacemaker system that drives automaticity in mammals are also active in the zebrafish, and that the effects of extra- and intracardiac control of heart rate seen in mammals are also present. Overall, these results, combined with previously published work, support the utility of the zebrafish as a novel experimental model for studies of SAN (patho-) physiological function.
Collapse
Affiliation(s)
- Matthew R. Stoyek
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
| | - Eilidh A. MacDonald
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Melissa Mantifel
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
| | - Jonathan S. Baillie
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
| | - Bailey M. Selig
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
| | - Roger P. Croll
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
| | - Frank M. Smith
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - T. Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
- *Correspondence: T. Alexander Quinn,
| |
Collapse
|
11
|
The effective use of blebbistatin to study the action potential of cardiac pacemaker cells of zebrafish (Danio rerio) during incremental warming. Curr Res Physiol 2022; 5:48-54. [PMID: 35128467 PMCID: PMC8803472 DOI: 10.1016/j.crphys.2022.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/12/2022] [Accepted: 01/12/2022] [Indexed: 12/11/2022] Open
Abstract
Blebbistatin potently inhibits actin-myosin interaction, preventing contractile activity of excitable cells including cardiac myocytes, despite electrical excitation of an action potential (AP). We collected intracellular microelectrode recordings of pacemaker cells located in the sinoatrial region (SAR) of the zebrafish heart at room temperature and during acute warming to investigate whether or not blebbistatin inhibition of contraction significantly alters pacemaker cell electrophysiology. Changes were evaluated based on 16 variables that characterized the AP waveform. None of these AP variables nor the spontaneous heart rate were significantly modified with the application of 10 μM blebbistatin when recordings were made at room temperature. Compared with the control group, the blebbistatin-treated group showed minor changes in the rate of spontaneous diastolic depolarization (P = 0.027) and the 50% and 80% repolarization (P = 0.008 and 0.010, respectively) in the 26°C–29°C temperature bin, but not at higher temperatures. These findings suggest that blebbistatin is an effective excitation-contraction uncoupler that does not appreciably affect APs generated in pacemaking cells of the SAR and can, therefore, be used in zebrafish cardiac studies. Blebbistatin uncouples excitation-contraction in zebrafish cardiomyocytes. Blebbistatin does not modify the pacemaker action potential variables. Temperature does not modify the effect of blebbistatin. First validation of the use of blebbistatin in adult fish. Methodology of intracellular microelectrode recording of zebrafish pacemaker cells.
Collapse
|
12
|
Baillie JS, Stoyek MR, Quinn TA. Seeing the Light: The Use of Zebrafish for Optogenetic Studies of the Heart. Front Physiol 2021; 12:748570. [PMID: 35002753 PMCID: PMC8733579 DOI: 10.3389/fphys.2021.748570] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
Optogenetics, involving the optical measurement and manipulation of cellular activity with genetically encoded light-sensitive proteins ("reporters" and "actuators"), is a powerful experimental technique for probing (patho-)physiological function. Originally developed as a tool for neuroscience, it has now been utilized in cardiac research for over a decade, providing novel insight into the electrophysiology of the healthy and diseased heart. Among the pioneering cardiac applications of optogenetic actuators were studies in zebrafish, which first demonstrated their use for precise spatiotemporal control of cardiac activity. Zebrafish were also adopted early as an experimental model for the use of optogenetic reporters, including genetically encoded voltage- and calcium-sensitive indicators. Beyond optogenetic studies, zebrafish are becoming an increasingly important tool for cardiac research, as they combine many of the advantages of integrative and reduced experimental models. The zebrafish has striking genetic and functional cardiac similarities to that of mammals, its genome is fully sequenced and can be modified using standard techniques, it has been used to recapitulate a variety of cardiac diseases, and it allows for high-throughput investigations. For optogenetic studies, zebrafish provide additional advantages, as the whole zebrafish heart can be visualized and interrogated in vivo in the transparent, externally developing embryo, and the relatively small adult heart allows for in situ cell-specific observation and control not possible in mammals. With the advent of increasingly sophisticated fluorescence imaging approaches and methods for spatially-resolved light stimulation in the heart, the zebrafish represents an experimental model with unrealized potential for cardiac optogenetic studies. In this review we summarize the use of zebrafish for optogenetic investigations in the heart, highlighting their specific advantages and limitations, and their potential for future cardiac research.
Collapse
Affiliation(s)
- Jonathan S. Baillie
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
| | - Matthew R. Stoyek
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
| | - T. Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
13
|
Stoyek MR, Hortells L, Quinn TA. From Mice to Mainframes: Experimental Models for Investigation of the Intracardiac Nervous System. J Cardiovasc Dev Dis 2021; 8:149. [PMID: 34821702 PMCID: PMC8620975 DOI: 10.3390/jcdd8110149] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 01/17/2023] Open
Abstract
The intracardiac nervous system (IcNS), sometimes referred to as the "little brain" of the heart, is involved in modulating many aspects of cardiac physiology. In recent years our fundamental understanding of autonomic control of the heart has drastically improved, and the IcNS is increasingly being viewed as a therapeutic target in cardiovascular disease. However, investigations of the physiology and specific roles of intracardiac neurons within the neural circuitry mediating cardiac control has been hampered by an incomplete knowledge of the anatomical organisation of the IcNS. A more thorough understanding of the IcNS is hoped to promote the development of new, highly targeted therapies to modulate IcNS activity in cardiovascular disease. In this paper, we first provide an overview of IcNS anatomy and function derived from experiments in mammals. We then provide descriptions of alternate experimental models for investigation of the IcNS, focusing on a non-mammalian model (zebrafish), neuron-cardiomyocyte co-cultures, and computational models to demonstrate how the similarity of the relevant processes in each model can help to further our understanding of the IcNS in health and disease.
Collapse
Affiliation(s)
- Matthew R. Stoyek
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS 15000, Canada;
| | - Luis Hortells
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg–Bad Krozingen, 79110 Freiburg, Germany;
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - T. Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS 15000, Canada;
- School of Biomedical Engineering, Dalhousie University, Halifax, NS 15000, Canada
| |
Collapse
|
14
|
Ye L, Rawls JF. Microbial influences on gut development and gut-brain communication. Development 2021; 148:dev194936. [PMID: 34758081 PMCID: PMC8627602 DOI: 10.1242/dev.194936] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 10/07/2021] [Indexed: 12/15/2022]
Abstract
The developmental programs that build and sustain animal forms also encode the capacity to sense and adapt to the microbial world within which they evolved. This is abundantly apparent in the development of the digestive tract, which typically harbors the densest microbial communities of the body. Here, we review studies in human, mouse, zebrafish and Drosophila that are revealing how the microbiota impacts the development of the gut and its communication with the nervous system, highlighting important implications for human and animal health.
Collapse
|
15
|
Abu Nahia K, Migdał M, Quinn TA, Poon KL, Łapiński M, Sulej A, Liu J, Mondal SS, Pawlak M, Bugajski Ł, Piwocka K, Brand T, Kohl P, Korzh V, Winata C. Genomic and physiological analyses of the zebrafish atrioventricular canal reveal molecular building blocks of the secondary pacemaker region. Cell Mol Life Sci 2021; 78:6669-6687. [PMID: 34557935 PMCID: PMC8558220 DOI: 10.1007/s00018-021-03939-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/06/2021] [Accepted: 09/10/2021] [Indexed: 01/06/2023]
Abstract
The atrioventricular canal (AVC) is the site where key structures responsible for functional division between heart regions are established, most importantly, the atrioventricular (AV) conduction system and cardiac valves. To elucidate the mechanism underlying AVC development and function, we utilized transgenic zebrafish line sqet31Et expressing EGFP in the AVC to isolate this cell population and profile its transcriptome at 48 and 72 hpf. The zebrafish AVC transcriptome exhibits hallmarks of mammalian AV node, including the expression of genes implicated in its development and those encoding connexins forming low conductance gap junctions. Transcriptome analysis uncovered protein-coding and noncoding transcripts enriched in AVC, which have not been previously associated with this structure, as well as dynamic expression of epithelial-to-mesenchymal transition markers and components of TGF-β, Notch, and Wnt signaling pathways likely reflecting ongoing AVC and valve development. Using transgenic line Tg(myl7:mermaid) encoding voltage-sensitive fluorescent protein, we show that abolishing the pacemaker-containing sinoatrial ring (SAR) through Isl1 loss of function resulted in spontaneous activation in the AVC region, suggesting that it possesses inherent automaticity although insufficient to replace the SAR. The SAR and AVC transcriptomes express partially overlapping species of ion channels and gap junction proteins, reflecting their distinct roles. Besides identifying conserved aspects between zebrafish and mammalian conduction systems, our results established molecular hallmarks of the developing AVC which underlies its role in structural and electrophysiological separation between heart chambers. This data constitutes a valuable resource for studying AVC development and function, and identification of novel candidate genes implicated in these processes.
Collapse
Affiliation(s)
- Karim Abu Nahia
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Maciej Migdał
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kar-Lai Poon
- Institute of Molecular and Cell Biology, 61 Biopolis Dr, Singapore , Singapore.,Developmental Dynamics, National Heart and Lung Institute, Imperial College London, London, UK
| | - Maciej Łapiński
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Agata Sulej
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Jiandong Liu
- McAllister Heart Institute, University of North Carolina, Chapel Hill, USA
| | - Shamba S Mondal
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Michał Pawlak
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | | | | | - Thomas Brand
- Developmental Dynamics, National Heart and Lung Institute, Imperial College London, London, UK
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Centre, Faculty of Medicine, and Faculty of Engineering, University of Freiburg, Freiburg im Breisgau, Germany
| | - Vladimir Korzh
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland.
| | - Cecilia Winata
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland.
| |
Collapse
|
16
|
Joyce W, Perry SF. Hif-1α is not required for the development of cardiac adrenergic control in zebrafish (Danio rerio). JOURNAL OF EXPERIMENTAL ZOOLOGY PART 2021; 335:623-631. [PMID: 34288573 DOI: 10.1002/jez.2507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/23/2021] [Accepted: 07/01/2021] [Indexed: 12/23/2022]
Abstract
Adrenergic regulation, acting via the sympathetic nervous system, provides a major mechanism to control cardiac function. It has recently been shown that hypoxia inducible factor-1α (Hif-1α) is necessary for normal development of sympathetic innervation and control of cardiac function in the mouse. To investigate whether this may represent a fundamental trait shared across vertebrates, we assessed adrenergic regulation of the heart in wild-type and Hif-1α knockout (hif-1α -/- ) zebrafish (Danio rerio). Wild-type and hif-1α -/- zebrafish larvae (aged 4 and 7 days postfertilisation) exhibited similar routine heart rates within a given age group, and β-adrenergic receptor blockade with propranolol universally reduced heart rate to comparable levels, indicating similar adrenergic tone in both genotypes. In adult fish, in vivo heart rate measured during anaesthesia was identical between genotypes. Treatment of spontaneously beating hearts in vitro with adrenaline revealed a similar positive chronotropic effect and similar maximum heart rates in both genotypes. Tyrosine hydroxylase immunohistochemistry with confocal microscopy demonstrated that the bulbus arteriosus (outflow tract of the teleost heart) of adult fish was particularly well innervated by sympathetic nerves, and nerve density (as a percentage of bulbus arteriosus area) was similar between wild-types and hif-1α -/- mutants. In summary, we did not find any evidence that adrenergic cardiac control was perturbed in larval or adult zebrafish lacking Hif-1α. We conclude that Hif-1α is not essential for the normal development of cardiovascular control or adult sympathetic cardiac innervation in zebrafish, although it is possible that it plays a redundant or auxiliary role.
Collapse
Affiliation(s)
- William Joyce
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada.,Department of Biology-Zoophysiology, Aarhus University, Aarhus C, Denmark
| | - Steve F Perry
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
17
|
Capillo G, Lauriano ER, Icardo JM, Siriyappagouder P, Kuciel M, Karapanagiotis S, Zaccone G, Fernandes JMO. Structural Identification of the Pacemaker Cells and Expression of Hyperpolarization-Activated Cyclic Nucleotide-Gated (HCN) Channels in the Heart of the Wild Atlantic Cod, Gadus morhua (Linnaeus, 1758). Int J Mol Sci 2021; 22:7539. [PMID: 34299159 PMCID: PMC8307021 DOI: 10.3390/ijms22147539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/05/2021] [Accepted: 07/12/2021] [Indexed: 11/16/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are proteins that contain highly conserved functional domains and sequence motifs that are correlated with their unique biophysical activities, to regulate cardiac pacemaker activity and synaptic transmission. These pacemaker proteins have been studied in mammalian species, but little is known now about their heart distribution in lower vertebrates and c-AMP modulation. Here, we characterized the pacemaker system in the heart of the wild Atlantic cod (Gadus morhua), with respect to primary pacemaker molecular markers. Special focus is given to the structural, ultrastructural and molecular characterization of the pacemaker domain, through the expression of HCN channel genes and the immunohistochemistry of HCN isoforms, including the location of intracardiac neurons that are adjacent to the sinoatrial region of the heart. Similarly to zebrafish and mammals, these neurons are immunoreactive to ChAT, VAChT and nNOS. It has been shown that cardiac pacemaking can be modulated by sympathetic and parasympathetic pathways, and the existence of intracardiac neurons projecting back to the central nervous system provide a plausible link between them.
Collapse
Affiliation(s)
- Gioele Capillo
- Department of Veterinary Sciences, Polo Universitario dell’Annunziata, University of Messina, 98168 Messina, Italy;
- Institute of Marine Biological Resources and Biotechnology—National Research Council (IRBIM, CNR), Spianata S. Raineri, 98122 Messina, Italy
| | - Eugenia R. Lauriano
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, 98168 Messina, Italy;
| | - Jose M. Icardo
- Department of Anatomy and Cell Biology, Poligono de Cazona, Faculty of Medicine, University of Cantabria, 39011 Santander, Spain
| | | | - Michal Kuciel
- Poison Information Centre, Department of Toxicology and Environmental Disease, Faculty of Medicine, Jagiellonian University, Kopernika 15, 30-501 Cracow, Poland;
| | - Stelios Karapanagiotis
- Faculty of Biosciences and Aquaculture, Nord University, 8026 Bodø, Norway; (P.S.); (S.K.)
| | - Giacomo Zaccone
- Department of Veterinary Sciences, Polo Universitario dell’Annunziata, University of Messina, 98168 Messina, Italy;
| | - Jorge M. O. Fernandes
- Faculty of Biosciences and Aquaculture, Nord University, 8026 Bodø, Norway; (P.S.); (S.K.)
| |
Collapse
|
18
|
Kolesnikova ЕE. Anatomical and Physiological Peculiarities
of the Heart in Jawless and Jawed Fish. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021020022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Hassinen M, Dzhumaniiazova I, Abramochkin DV, Vornanen M. Ionic basis of atrioventricular conduction: ion channel expression and sarcolemmal ion currents of the atrioventricular canal of the rainbow trout (Oncorhynchus mykiss) heart. J Comp Physiol B 2021; 191:327-346. [PMID: 33575867 PMCID: PMC7895799 DOI: 10.1007/s00360-021-01344-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 12/15/2020] [Accepted: 01/18/2021] [Indexed: 12/17/2022]
Abstract
Atrioventricular (AV) nodal tissue synchronizes activities of atria and ventricles of the vertebrate heart and is also a potential site of cardiac arrhythmia, e.g., under acute heat stress. Since ion channel composition and ion currents of the fish AV canal have not been previously studied, we measured major cation currents and transcript expression of ion channels in rainbow trout (Oncorhynchus mykiss) AV tissue. Both ion current densities and expression of ion channel transcripts indicate that the fish AV canal has a characteristic electrophysiological phenotype that differs from those of sinoatrial tissue, atrium and ventricle. Two types of cardiomyocytes were distinguished electrophysiologically in trout AV nodal tissue: the one (transitional cell) is functionally intermediate between working atrial/ventricular myocytes and the other (AV nodal cell) has a less negative resting membrane potential than atrial and ventricular myocytes and is a more similar to the sinoatrial nodal cells in ion channel composition. The AV nodal cells are characterized by a small or non-existent inward rectifier potassium current (IK1), low density of fast sodium current (INa) and relatively high expression of T-type calcium channels (CACNA3.1). Pacemaker channel (HCN4 and HCN2) transcripts were expressed in the AV nodal tissue but If current was not found in enzymatically isolated nodal myocytes. The electrophysiological properties of the rainbow trout nodal cells are appropriate for a slow rate of action potential conduction (small INa) and a moderate propensity for pacemaking activity (absence of IK1).
Collapse
Affiliation(s)
- Minna Hassinen
- Department of Environmental and Biological Sciences, University of Eastern Finland, P.O. Box 111, 80101, Joensuu, Finland
| | - Irina Dzhumaniiazova
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia
| | - Denis V Abramochkin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia.,Laboratory of Cardiac Electrophysiology, National Medical Research Center for Cardiology, Moscow, Russia.,Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Matti Vornanen
- Department of Environmental and Biological Sciences, University of Eastern Finland, P.O. Box 111, 80101, Joensuu, Finland.
| |
Collapse
|
20
|
Simpson KE, Venkateshappa R, Pang ZK, Faizi S, Tibbits GF, Claydon TW. Utility of Zebrafish Models of Acquired and Inherited Long QT Syndrome. Front Physiol 2021; 11:624129. [PMID: 33519527 PMCID: PMC7844309 DOI: 10.3389/fphys.2020.624129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/21/2020] [Indexed: 01/12/2023] Open
Abstract
Long-QT Syndrome (LQTS) is a cardiac electrical disorder, distinguished by irregular heart rates and sudden death. Accounting for ∼40% of cases, LQTS Type 2 (LQTS2), is caused by defects in the Kv11.1 (hERG) potassium channel that is critical for cardiac repolarization. Drug block of hERG channels or dysfunctional channel variants can result in acquired or inherited LQTS2, respectively, which are typified by delayed repolarization and predisposition to lethal arrhythmia. As such, there is significant interest in clear identification of drugs and channel variants that produce clinically meaningful perturbation of hERG channel function. While toxicological screening of hERG channels, and phenotypic assessment of inherited channel variants in heterologous systems is now commonplace, affordable, efficient, and insightful whole organ models for acquired and inherited LQTS2 are lacking. Recent work has shown that zebrafish provide a viable in vivo or whole organ model of cardiac electrophysiology. Characterization of cardiac ion currents and toxicological screening work in intact embryos, as well as adult whole hearts, has demonstrated the utility of the zebrafish model to contribute to the development of therapeutics that lack hERG-blocking off-target effects. Moreover, forward and reverse genetic approaches show zebrafish as a tractable model in which LQTS2 can be studied. With the development of new tools and technologies, zebrafish lines carrying precise channel variants associated with LQTS2 have recently begun to be generated and explored. In this review, we discuss the present knowledge and questions raised related to the use of zebrafish as models of acquired and inherited LQTS2. We focus discussion, in particular, on developments in precise gene-editing approaches in zebrafish to create whole heart inherited LQTS2 models and evidence that zebrafish hearts can be used to study arrhythmogenicity and to identify potential anti-arrhythmic compounds.
Collapse
Affiliation(s)
- Kyle E. Simpson
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Ravichandra Venkateshappa
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Zhao Kai Pang
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Shoaib Faizi
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Glen F. Tibbits
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- Department of Cardiovascular Science, British Columbia Children’s Hospital, Vancouver, BC, Canada
| | - Tom W. Claydon
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
21
|
Echeazarra L, Hortigón-Vinagre MP, Casis O, Gallego M. Adult and Developing Zebrafish as Suitable Models for Cardiac Electrophysiology and Pathology in Research and Industry. Front Physiol 2021; 11:607860. [PMID: 33519514 PMCID: PMC7838705 DOI: 10.3389/fphys.2020.607860] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022] Open
Abstract
The electrophysiological behavior of the zebrafish heart is very similar to that of the human heart. In fact, most of the genes that codify the channels and regulatory proteins required for human cardiac function have their orthologs in the zebrafish. The high fecundity, small size, and easy handling make the zebrafish embryos/larvae an interesting candidate to perform whole animal experiments within a plate, offering a reliable and low-cost alternative to replace rodents and larger mammals for the study of cardiac physiology and pathology. The employment of zebrafish embryos/larvae has widened from basic science to industry, being of particular interest for pharmacology studies, since the zebrafish embryo/larva is able to recapitulate a complete and integrated view of cardiac physiology, missed in cell culture. As in the human heart, IKr is the dominant repolarizing current and it is functional as early as 48 h post fertilization. Finally, genome editing techniques such as CRISPR/Cas9 facilitate the humanization of zebrafish embryos/larvae. These techniques allow one to replace zebrafish genes by their human orthologs, making humanized zebrafish embryos/larvae the most promising in vitro model, since it allows the recreation of human-organ-like environment, which is especially necessary in cardiac studies due to the implication of dynamic factors, electrical communication, and the paracrine signals in cardiac function.
Collapse
Affiliation(s)
- Leyre Echeazarra
- Departamento de Fisiología, Facultad de Farmacia, Universidad del País Vasco UPV/EHU, Vitoria-Gasteiz, Spain
| | - Maria Pura Hortigón-Vinagre
- Departamento de Bioquímica y Biología Molecular y Genética>, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Oscar Casis
- Departamento de Fisiología, Facultad de Farmacia, Universidad del País Vasco UPV/EHU, Vitoria-Gasteiz, Spain
| | - Mónica Gallego
- Departamento de Fisiología, Facultad de Farmacia, Universidad del País Vasco UPV/EHU, Vitoria-Gasteiz, Spain
| |
Collapse
|
22
|
Fedele L, Brand T. The Intrinsic Cardiac Nervous System and Its Role in Cardiac Pacemaking and Conduction. J Cardiovasc Dev Dis 2020; 7:jcdd7040054. [PMID: 33255284 PMCID: PMC7712215 DOI: 10.3390/jcdd7040054] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/11/2022] Open
Abstract
The cardiac autonomic nervous system (CANS) plays a key role for the regulation of cardiac activity with its dysregulation being involved in various heart diseases, such as cardiac arrhythmias. The CANS comprises the extrinsic and intrinsic innervation of the heart. The intrinsic cardiac nervous system (ICNS) includes the network of the intracardiac ganglia and interconnecting neurons. The cardiac ganglia contribute to the tight modulation of cardiac electrophysiology, working as a local hub integrating the inputs of the extrinsic innervation and the ICNS. A better understanding of the role of the ICNS for the modulation of the cardiac conduction system will be crucial for targeted therapies of various arrhythmias. We describe the embryonic development, anatomy, and physiology of the ICNS. By correlating the topography of the intracardiac neurons with what is known regarding their biophysical and neurochemical properties, we outline their physiological role in the control of pacemaker activity of the sinoatrial and atrioventricular nodes. We conclude by highlighting cardiac disorders with a putative involvement of the ICNS and outline open questions that need to be addressed in order to better understand the physiology and pathophysiology of the ICNS.
Collapse
Affiliation(s)
- Laura Fedele
- Correspondence: (L.F.); (T.B.); Tel.: +44-(0)-207-594-6531 (L.F.); +44-(0)-207-594-8744 (T.B.)
| | - Thomas Brand
- Correspondence: (L.F.); (T.B.); Tel.: +44-(0)-207-594-6531 (L.F.); +44-(0)-207-594-8744 (T.B.)
| |
Collapse
|
23
|
Sutcliffe RL, Li S, Gilbert MJH, Schulte PM, Miller KM, Farrell AP. A rapid intrinsic heart rate resetting response with thermal acclimation in rainbow trout, Oncorhynchus mykiss. J Exp Biol 2020; 223:jeb215210. [PMID: 32345705 PMCID: PMC7328139 DOI: 10.1242/jeb.215210] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 04/17/2020] [Indexed: 01/01/2023]
Abstract
We examined cardiac pacemaker rate resetting in rainbow trout following a reciprocal temperature transfer. In the original experiment, performed in winter, 4°C-acclimated fish transferred to 12°C reset intrinsic heart rate after just 1 h (from 56.8±1.2 to 50.8±1.5 beats min-1); 12°C-acclimated fish transferred to 4°C reset intrinsic heart rate after 8 h (from 33.4±0.7 to 37.7±1.2 beats min-1). However, in a replicate experiment, performed in the summer using a different brood year, intrinsic heart rate was not reset, even after 10 weeks at a new temperature. Using this serendipitous opportunity, we compared mRNA expression changes of a suite of proteins in sinoatrial node (SAN), atrial and ventricular tissues after both 1 h and longer than 3 weeks for both experimental acclimation groups to identify those changes only associated with pacemaker rate resetting. Of the changes in mRNA expression occurring after more than 3 weeks of warm acclimation and associated with pacemaker rate resetting, we observed downregulation of NKA α1c in the atrium and ventricle, and upregulation of HCN1 in the ventricle. However, in the SAN there were no mRNA expression changes unique to the fish with pacemaker rate resetting after either 1 h or 3 weeks of warm acclimation. Thus, despite identifying changes in mRNA expression of contractile cardiac tissues, there was an absence of changes in mRNA expression directly involved with the initial, rapid pacemaker rate resetting with warm acclimation. Importantly, pacemaker rate resetting with thermal acclimation does not always occur in rainbow trout.
Collapse
Affiliation(s)
- Rachel L Sutcliffe
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada, V6T 1Z4
| | - Shaorong Li
- Pacific Biological Station, Fisheries and Oceans, Nanaimo, BC, Canada, V9T 6N7
| | - Matthew J H Gilbert
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada, V6T 1Z4
| | - Patricia M Schulte
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada, V6T 1Z4
| | - Kristi M Miller
- Pacific Biological Station, Fisheries and Oceans, Nanaimo, BC, Canada, V9T 6N7
| | - Anthony P Farrell
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada, V6T 1Z4
| |
Collapse
|
24
|
Yan J, Li H, Bu H, Jiao K, Zhang AX, Le T, Cao H, Li Y, Ding Y, Xu X. Aging-associated sinus arrest and sick sinus syndrome in adult zebrafish. PLoS One 2020; 15:e0232457. [PMID: 32401822 PMCID: PMC7219707 DOI: 10.1371/journal.pone.0232457] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 04/15/2020] [Indexed: 12/24/2022] Open
Abstract
Because of its powerful genetics, the adult zebrafish has been increasingly used for studying cardiovascular diseases. Considering its heart rate of ~100 beats per minute at ambient temperature, which is very close to human, we assessed the use of this vertebrate animal for modeling heart rhythm disorders such as sinus arrest (SA) and sick sinus syndrome (SSS). We firstly optimized a protocol to measure electrocardiogram in adult zebrafish. We determined the location of the probes, implemented an open-chest microsurgery procedure, measured the effects of temperature, and determined appropriate anesthesia dose and time. We then proposed an PP interval of more than 1.5 seconds as an arbitrary criterion to define an SA episode in an adult fish at ambient temperature, based on comparison between the current definition of an SA episode in humans and our studies of candidate SA episodes in aged wild-type fish and Tg(SCN5A-D1275N) fish (a fish model for inherited SSS). With this criterion, a subpopulation of about 5% wild-type fish can be considered to have SA episodes, and this percentage significantly increases to about 25% in 3-year-old fish. In response to atropine, this subpopulation has both common SSS phenotypic traits that are shared with the Tg(SCN5A-D1275N) model, such as bradycardia; and unique SSS phenotypic traits, such as increased QRS/P ratio and chronotropic incompetence. In summary, this study defined baseline SA and SSS in adult zebrafish and underscored use of the zebrafish as an alternative model to study aging-associated SSS.
Collapse
Affiliation(s)
- Jianhua Yan
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- Division of Cardiology, Xinhua Hospital Affiliated To Shanghai Jiaotong University School Of Medicine, Shanghai, China
| | - Hongsong Li
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Haisong Bu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kunli Jiao
- Division of Cardiology, Xinhua Hospital Affiliated To Shanghai Jiaotong University School Of Medicine, Shanghai, China
| | - Alex X. Zhang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Tai Le
- Department of Electrical Engineering and Computer Science, UC Irvine, Irvine, California
| | - Hung Cao
- Department of Electrical Engineering and Computer Science, UC Irvine, Irvine, California
- Department of Biomedical Engineering, UC Irvine, Irvine, California
| | - Yigang Li
- Division of Cardiology, Xinhua Hospital Affiliated To Shanghai Jiaotong University School Of Medicine, Shanghai, China
| | - Yonghe Ding
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| |
Collapse
|
25
|
Shrestha R, Lieberth J, Tillman S, Natalizio J, Bloomekatz J. Using Zebrafish to Analyze the Genetic and Environmental Etiologies of Congenital Heart Defects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:189-223. [PMID: 32304074 DOI: 10.1007/978-981-15-2389-2_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Congenital heart defects (CHDs) are among the most common human birth defects. However, the etiology of a large proportion of CHDs remains undefined. Studies identifying the molecular and cellular mechanisms that underlie cardiac development have been critical to elucidating the origin of CHDs. Building upon this knowledge to understand the pathogenesis of CHDs requires examining how genetic or environmental stress changes normal cardiac development. Due to strong molecular conservation to humans and unique technical advantages, studies using zebrafish have elucidated both fundamental principles of cardiac development and have been used to create cardiac disease models. In this chapter we examine the unique toolset available to zebrafish researchers and how those tools are used to interrogate the genetic and environmental contributions to CHDs.
Collapse
Affiliation(s)
- Rabina Shrestha
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Jaret Lieberth
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Savanna Tillman
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Joseph Natalizio
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | | |
Collapse
|
26
|
MacDonald EA, Rose RA, Quinn TA. Neurohumoral Control of Sinoatrial Node Activity and Heart Rate: Insight From Experimental Models and Findings From Humans. Front Physiol 2020; 11:170. [PMID: 32194439 PMCID: PMC7063087 DOI: 10.3389/fphys.2020.00170] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 02/13/2020] [Indexed: 12/22/2022] Open
Abstract
The sinoatrial node is perhaps one of the most important tissues in the entire body: it is the natural pacemaker of the heart, making it responsible for initiating each-and-every normal heartbeat. As such, its activity is heavily controlled, allowing heart rate to rapidly adapt to changes in physiological demand. Control of sinoatrial node activity, however, is complex, occurring through the autonomic nervous system and various circulating and locally released factors. In this review we discuss the coupled-clock pacemaker system and how its manipulation by neurohumoral signaling alters heart rate, considering the multitude of canonical and non-canonical agents that are known to modulate sinoatrial node activity. For each, we discuss the principal receptors involved and known intracellular signaling and protein targets, highlighting gaps in our knowledge and understanding from experimental models and human studies that represent areas for future research.
Collapse
Affiliation(s)
- Eilidh A. MacDonald
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
| | - Robert A. Rose
- Cumming School of Medicine, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | - T. Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
27
|
The ventral peptidergic system of the adult ascidian Ciona robusta (Ciona intestinalis Type A) insights from a transgenic animal model. Sci Rep 2020; 10:1892. [PMID: 32024913 PMCID: PMC7002689 DOI: 10.1038/s41598-020-58884-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
Ascidians are the sister group of vertebrates and occupy a critical position in explorations of the evolution of the endocrine and nervous systems of chordates. Here, we describe the complete ventral peptidergic system in adult transgenic Ciona robusta (Ciona intestinalis Type A) which expresses the Kaede reporter gene driven by the prohormone convertase 2 (PC2) gene promoter. Numerous PC2 promoter-driven fluorescent (Kaede-positive) non-neural cells were distributed in the blood sinus located at the anterior end of the pharynx, suggesting the acquisition of a peptidergic circulatory system in Ciona. Kaede-positive ciliated columnar cells, rounded cells, and tall ciliated cells were observed in the alimentary organs, including the endostyle, pharynx, esophagus, stomach, and intestine, suggesting that digestive functions are regulated by multiple peptidergic systems. In the heart, Kaede-positive neurons were located in the ring-shaped plexus at both ends of the myocardium. Nerve fiber-like tracts ran along the raphe and appeared to be connected with the plexuses. Such unique structures suggest a role for the peptidergic system in cardiac function. Collectively, the present anatomic analysis revealed the major framework of the ventral peptidergic system of adult Ciona, which could facilitate investigations of peptidergic regulation of the pharynx, endostyle, alimentary tissues, and heart.
Collapse
|
28
|
Physiological phenotyping of the adult zebrafish heart. Mar Genomics 2020; 49:100701. [DOI: 10.1016/j.margen.2019.100701] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 08/11/2019] [Accepted: 08/13/2019] [Indexed: 12/27/2022]
|
29
|
Kopton RA, Baillie JS, Rafferty SA, Moss R, Zgierski-Johnston CM, Prykhozhij SV, Stoyek MR, Smith FM, Kohl P, Quinn TA, Schneider-Warme F. Cardiac Electrophysiological Effects of Light-Activated Chloride Channels. Front Physiol 2018; 9:1806. [PMID: 30618818 PMCID: PMC6304430 DOI: 10.3389/fphys.2018.01806] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 11/30/2018] [Indexed: 12/17/2022] Open
Abstract
During the last decade, optogenetics has emerged as a paradigm-shifting technique to monitor and steer the behavior of specific cell types in excitable tissues, including the heart. Activation of cation-conducting channelrhodopsins (ChR) leads to membrane depolarization, allowing one to effectively trigger action potentials (AP) in cardiomyocytes. In contrast, the quest for optogenetic tools for hyperpolarization-induced inhibition of AP generation has remained challenging. The green-light activated ChR from Guillardia theta (GtACR1) mediates Cl--driven photocurrents that have been shown to silence AP generation in different types of neurons. It has been suggested, therefore, to be a suitable tool for inhibition of cardiomyocyte activity. Using single-cell electrophysiological recordings and contraction tracking, as well as intracellular microelectrode recordings and in vivo optical recordings of whole hearts, we find that GtACR1 activation by prolonged illumination arrests cardiac cells in a depolarized state, thus inhibiting re-excitation. In line with this, GtACR1 activation by transient light pulses elicits AP in rabbit isolated cardiomyocytes and in spontaneously beating intact hearts of zebrafish. Our results show that GtACR1 inhibition of AP generation is caused by cell depolarization. While this does not address the need for optogenetic silencing through physiological means (i.e., hyperpolarization), GtACR1 is a potentially attractive tool for activating cardiomyocytes by transient light-induced depolarization.
Collapse
Affiliation(s)
- Ramona A Kopton
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg-Bad Krozingen Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine University of Freiburg, Freiburg, Germany.,Faculty of Biology University of Freiburg, Freiburg, Germany
| | - Jonathan S Baillie
- Department of Physiology and Biophysics, Dalhousie University Halifax, NS, Canada
| | - Sara A Rafferty
- Department of Physiology and Biophysics, Dalhousie University Halifax, NS, Canada
| | - Robin Moss
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg-Bad Krozingen Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - Callum M Zgierski-Johnston
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg-Bad Krozingen Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine University of Freiburg, Freiburg, Germany
| | | | - Matthew R Stoyek
- Department of Physiology and Biophysics, Dalhousie University Halifax, NS, Canada
| | - Frank M Smith
- Department of Medical Neuroscience, Dalhousie University Halifax, NS, Canada
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg-Bad Krozingen Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University Halifax, NS, Canada.,School of Biomedical Engineering, Dalhousie University Halifax, NS, Canada
| | - Franziska Schneider-Warme
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg-Bad Krozingen Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine University of Freiburg, Freiburg, Germany
| |
Collapse
|
30
|
Ravens U. Ionic basis of cardiac electrophysiology in zebrafish compared to human hearts. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 138:38-44. [DOI: 10.1016/j.pbiomolbio.2018.06.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/23/2018] [Accepted: 06/15/2018] [Indexed: 12/14/2022]
|
31
|
Stoyek MR, Rog-Zielinska EA, Quinn TA. Age-associated changes in electrical function of the zebrafish heart. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 138:91-104. [DOI: 10.1016/j.pbiomolbio.2018.07.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/24/2018] [Accepted: 07/25/2018] [Indexed: 12/17/2022]
|
32
|
Zhang H, Dvornikov AV, Huttner IG, Ma X, Santiago CF, Fatkin D, Xu X. A Langendorff-like system to quantify cardiac pump function in adult zebrafish. Dis Model Mech 2018; 11:dmm.034819. [PMID: 30012855 PMCID: PMC6177000 DOI: 10.1242/dmm.034819] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/10/2018] [Indexed: 12/22/2022] Open
Abstract
Zebrafish are increasingly used as a vertebrate model to study human cardiovascular disorders. Although heart structure and function are readily visualized in zebrafish embryos because of their optical transparency, the lack of effective tools for evaluating the hearts of older, nontransparent fish has been a major limiting factor. The recent development of high-frequency echocardiography has been an important advance for in vivo cardiac assessment, but it necessitates anesthesia and has limited ability to study acute interventions. We report the development of an alternative experimental ex vivo technique for quantifying heart size and function that resembles the Langendorff heart preparations that have been widely used in mammalian models. Dissected adult zebrafish hearts were perfused with a calcium-containing buffer, and a beat frequency was maintained with electrical stimulation. The impact of pacing frequency, flow rate and perfusate calcium concentration on ventricular performance (including end-diastolic and end-systolic volumes, ejection fraction, radial strain, and maximal velocities of shortening and relaxation) were evaluated and optimal conditions defined. We determined the effects of age on heart function in wild-type male and female zebrafish, and successfully detected hypercontractile and hypocontractile responses after adrenergic stimulation or doxorubicin treatment, respectively. Good correlations were found between indices of cardiac contractility obtained with high-frequency echocardiography and with the ex vivo technique in a subset of fish studied with both methods. The ex vivo beating heart preparation is a valuable addition to the cardiac function tool kit that will expand the use of adult zebrafish for cardiovascular research.
Collapse
Affiliation(s)
- Hong Zhang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55902, USA.,Cardiovascular Surgery Department, the Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Alexey V Dvornikov
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55902, USA
| | - Inken G Huttner
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Xiao Ma
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55902, USA.,Clinical and Translational Sciences Track, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine and Science, Rochester, MN 55092, USA
| | - Celine F Santiago
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Diane Fatkin
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.,Cardiology Department, St. Vincent's Hospital, Sydney, NSW 2010, Australia
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55902, USA
| |
Collapse
|
33
|
Cloney K, Steele SL, Stoyek MR, Croll RP, Smith FM, Prykhozhij SV, Brown MM, Midgen C, Blake K, Berman JN. Etiology and functional validation of gastrointestinal motility dysfunction in a zebrafish model of CHARGE syndrome. FEBS J 2018; 285:2125-2140. [PMID: 29660852 DOI: 10.1111/febs.14473] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 03/17/2018] [Accepted: 04/09/2018] [Indexed: 12/21/2022]
Abstract
CHARGE syndrome is linked to autosomal-dominant mutations in the CHD7 gene and results in a number of physiological and structural abnormalities, including heart defects, hearing and vision loss, and gastrointestinal (GI) problems. Of these challenges, GI problems have a profound impact throughout an individual's life, resulting in increased morbidity and mortality. A homolog of CHD7 has been identified in the zebrafish, the loss of which recapitulates many of the features of the human disease. Using a morpholino chd7 knockdown model complemented by a chd7 null mutant zebrafish line, we examined GI structure, innervation, and motility in larval zebrafish. Loss of chd7 resulted in physically smaller GI tracts with normal epithelial and muscular histology, but decreased and disorganized vagal projections, particularly in the foregut. chd7 morphant larvae had significantly less ability to empty their GI tract of gavaged fluorescent beads, and this condition was only minimally improved by the prokinetic agents, domperidone and erythromycin, in keeping with mixed responses to these agents in patients with CHARGE syndrome. The conserved genetics and transparency of the zebrafish have provided new insights into the consequences of chd7 gene dysfunction on the GI system and cranial nerve patterning. These findings highlight the opportunity of the zebrafish to serve as a preclinical model for studying compounds that may improve GI motility in individuals with CHARGE syndrome.
Collapse
Affiliation(s)
- Kellie Cloney
- Faculty of Medicine, Dalhousie University, Halifax, Canada
| | - Shelby L Steele
- Department of Pediatrics, Dalhousie University, Halifax, Canada
| | - Matthew R Stoyek
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Roger P Croll
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Frank M Smith
- Department of Medical Neuroscience, Dalhousie University, Halifax, Canada
| | | | - Mary M Brown
- Departments of Pediatrics and Obstetrics and Gynaecology, Dalhousie University, Halifax, Canada
| | - Craig Midgen
- Department of Pathology, Dalhousie University, Halifax, Canada
| | - Kim Blake
- Faculty of Medicine, Dalhousie University, Halifax, Canada.,Department of Pediatrics, Dalhousie University, Halifax, Canada
| | - Jason N Berman
- Department of Pediatrics, Dalhousie University, Halifax, Canada.,Department of Pathology, Dalhousie University, Halifax, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, Canada
| |
Collapse
|
34
|
Burkhard SB, Bakkers J. Spatially resolved RNA-sequencing of the embryonic heart identifies a role for Wnt/β-catenin signaling in autonomic control of heart rate. eLife 2018; 7:31515. [PMID: 29400650 PMCID: PMC5815850 DOI: 10.7554/elife.31515] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 02/03/2018] [Indexed: 11/13/2022] Open
Abstract
Development of specialized cells and structures in the heart is regulated by spatially -restricted molecular pathways. Disruptions in these pathways can cause severe congenital cardiac malformations or functional defects. To better understand these pathways and how they regulate cardiac development we used tomo-seq, combining high-throughput RNA-sequencing with tissue-sectioning, to establish a genome-wide expression dataset with high spatial resolution for the developing zebrafish heart. Analysis of the dataset revealed over 1100 genes differentially expressed in sub-compartments. Pacemaker cells in the sinoatrial region induce heart contractions, but little is known about the mechanisms underlying their development. Using our transcriptome map, we identified spatially restricted Wnt/β-catenin signaling activity in pacemaker cells, which was controlled by Islet-1 activity. Moreover, Wnt/β-catenin signaling controls heart rate by regulating pacemaker cellular response to parasympathetic stimuli. Thus, this high-resolution transcriptome map incorporating all cell types in the embryonic heart can expose spatially restricted molecular pathways critical for specific cardiac functions.
Collapse
Affiliation(s)
- Silja Barbara Burkhard
- Hubrecht Institute-KNAW, Utrecht, Netherlands.,University Medical Center Utrecht, Utrecht, Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW, Utrecht, Netherlands.,University Medical Center Utrecht, Utrecht, Netherlands.,Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
35
|
Stoyek MR, Schmidt MK, Wilfart FM, Croll RP, Smith FM. The in vitro zebrafish heart as a model to investigate the chronotropic effects of vapor anesthetics. Am J Physiol Regul Integr Comp Physiol 2017; 313:R669-R679. [PMID: 28877873 DOI: 10.1152/ajpregu.00467.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 08/15/2017] [Accepted: 09/05/2017] [Indexed: 11/22/2022]
Abstract
In addition to their intended clinical actions, all general anesthetic agents in common use have detrimental intrasurgical and postsurgical side effects on organs and systems, including the heart. The major cardiac side effect of anesthesia is bradycardia, which increases the probability of insufficient systemic perfusion during surgery. These side effects also occur in all vertebrate species so far examined, but the underlying mechanisms are not clear. The zebrafish heart is a powerful model for studying cardiac electrophysiology, employing the same pacemaker system and neural control as do mammalian hearts. In this study, isolated zebrafish hearts were significantly bradycardic during exposure to the vapor anesthetics sevoflurane (SEVO), desflurane (DES), and isoflurane (ISO). Bradycardia induced by DES and ISO continued during pharmacological blockade of the intracardiac portion of the autonomic nervous system, but the chronotropic effect of SEVO was eliminated during blockade. Bradycardia evoked by vagosympathetic nerve stimulation was augmented during DES and ISO exposure; nerve stimulation during SEVO exposure had no effect. Together, these results support the hypothesis that the cardiac chronotropic effect of SEVO occurs via a neurally mediated mechanism, while DES and ISO act directly upon cardiac pacemaker cells via an as yet unknown mechanism.
Collapse
Affiliation(s)
- Matthew R Stoyek
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Michael K Schmidt
- Department of Anesthesia, Pain Management and Perioperative Care, Dalhousie University, Halifax, Nova Scotia, Canada; and
| | - Florentin M Wilfart
- Department of Anesthesia, Pain Management and Perioperative Care, Dalhousie University, Halifax, Nova Scotia, Canada; and
| | - Roger P Croll
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Frank M Smith
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
36
|
Stoyek MR, Jonz MG, Smith FM, Croll RP. Distribution and chronotropic effects of serotonin in the zebrafish heart. Auton Neurosci 2017; 206:43-50. [DOI: 10.1016/j.autneu.2017.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 05/03/2017] [Accepted: 07/17/2017] [Indexed: 01/23/2023]
|
37
|
MacDonald EA, Stoyek MR, Rose RA, Quinn TA. Intrinsic regulation of sinoatrial node function and the zebrafish as a model of stretch effects on pacemaking. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017; 130:198-211. [PMID: 28743586 DOI: 10.1016/j.pbiomolbio.2017.07.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 07/17/2017] [Accepted: 07/21/2017] [Indexed: 12/18/2022]
Abstract
Excitation of the heart occurs in a specialised region known as the sinoatrial node (SAN). Tight regulation of SAN function is essential for the maintenance of normal heart rhythm and the response to (patho-)physiological changes. The SAN is regulated by extrinsic (central nervous system) and intrinsic (neurons, peptides, mechanics) factors. The positive chronotropic response to stretch in particular is essential for beat-by-beat adaptation to changes in hemodynamic load. Yet, the mechanism of this stretch response is unknown, due in part to the lack of an appropriate experimental model for targeted investigations. We have been investigating the zebrafish as a model for the study of intrinsic regulation of SAN function. In this paper, we first briefly review current knowledge of the principal components of extrinsic and intrinsic SAN regulation, derived primarily from experiments in mammals, followed by a description of the zebrafish as a novel experimental model for studies of intrinsic SAN regulation. This mini-review is followed by an original investigation of the response of the zebrafish isolated SAN to controlled stretch. Stretch causes an immediate and continuous increase in beating rate in the zebrafish isolated SAN. This increase reaches a maximum part way through a period of sustained stretch, with the total change dependent on the magnitude and direction of stretch. This is comparable to what occurs in isolated SAN from most mammals (including human), suggesting that the zebrafish is a novel experimental model for the study of mechanisms involved in the intrinsic regulation of SAN function by mechanical effects.
Collapse
Affiliation(s)
- Eilidh A MacDonald
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Matthew R Stoyek
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Robert A Rose
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada; School of Biomedical Engineering, Dalhousie University, Halifax, Canada.
| |
Collapse
|
38
|
Gut P, Reischauer S, Stainier DYR, Arnaout R. LITTLE FISH, BIG DATA: ZEBRAFISH AS A MODEL FOR CARDIOVASCULAR AND METABOLIC DISEASE. Physiol Rev 2017; 97:889-938. [PMID: 28468832 PMCID: PMC5817164 DOI: 10.1152/physrev.00038.2016] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 12/17/2022] Open
Abstract
The burden of cardiovascular and metabolic diseases worldwide is staggering. The emergence of systems approaches in biology promises new therapies, faster and cheaper diagnostics, and personalized medicine. However, a profound understanding of pathogenic mechanisms at the cellular and molecular levels remains a fundamental requirement for discovery and therapeutics. Animal models of human disease are cornerstones of drug discovery as they allow identification of novel pharmacological targets by linking gene function with pathogenesis. The zebrafish model has been used for decades to study development and pathophysiology. More than ever, the specific strengths of the zebrafish model make it a prime partner in an age of discovery transformed by big-data approaches to genomics and disease. Zebrafish share a largely conserved physiology and anatomy with mammals. They allow a wide range of genetic manipulations, including the latest genome engineering approaches. They can be bred and studied with remarkable speed, enabling a range of large-scale phenotypic screens. Finally, zebrafish demonstrate an impressive regenerative capacity scientists hope to unlock in humans. Here, we provide a comprehensive guide on applications of zebrafish to investigate cardiovascular and metabolic diseases. We delineate advantages and limitations of zebrafish models of human disease and summarize their most significant contributions to understanding disease progression to date.
Collapse
Affiliation(s)
- Philipp Gut
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Sven Reischauer
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Didier Y R Stainier
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Rima Arnaout
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| |
Collapse
|
39
|
Harrington JK, Sorabella R, Tercek A, Isler JR, Targoff KL. Nkx2.5 is essential to establish normal heart rate variability in the zebrafish embryo. Am J Physiol Regul Integr Comp Physiol 2017; 313:R265-R271. [PMID: 28615160 DOI: 10.1152/ajpregu.00223.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 12/11/2022]
Abstract
Heart rate variability (HRV) has become an important clinical marker of cardiovascular health and a research measure for the study of the cardiac conduction system and its autonomic controls. While the zebrafish (Danio rerio) is an ideal vertebrate model for understanding heart development, HRV has only recently been investigated in this system. We have previously demonstrated that nkx2.5 and nkx2.7, two homologues of Nkx2-5 expressed in zebrafish cardiomyocytes, play vital roles in maintaining cardiac chamber-specific characteristics. Given observed defects in ventricular and atrial chamber identities in nkx2.5-/- embryos coupled with conduction system abnormalities in murine models of Nkx2.5 insufficiency, we postulated that reduced HRV would serve as a marker of poor cardiac health in nkx2.5 mutants and in other zebrafish models of human congenital heart disease. Using live video image acquisition, we derived beat-to-beat intervals to compare HRV in wild-type and nkx2.5-/- embryos. Our data illustrate that the nkx2.5 loss-of-function model exhibits increased heart rate and decreased HRV when compared with wild type during embryogenesis. These findings validate HRV analysis as a useful quantitative tool for assessment of cardiac health in zebrafish and underscore the importance of nkx2.5 in maintaining normal heart rate and HRV during early conduction system development.
Collapse
Affiliation(s)
- Jamie K Harrington
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Robert Sorabella
- Division of Cardiothoracic Surgery, Department of Surgery, College of Physicians and Surgeons, Columbia University, New York, New York; and
| | - Abigail Tercek
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Joseph R Isler
- Division of Neonatology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Kimara L Targoff
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York;
| |
Collapse
|
40
|
Burkhard S, van Eif V, Garric L, Christoffels VM, Bakkers J. On the Evolution of the Cardiac Pacemaker. J Cardiovasc Dev Dis 2017; 4:jcdd4020004. [PMID: 29367536 PMCID: PMC5715705 DOI: 10.3390/jcdd4020004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 04/18/2017] [Accepted: 04/24/2017] [Indexed: 01/26/2023] Open
Abstract
The rhythmic contraction of the heart is initiated and controlled by an intrinsic pacemaker system. Cardiac contractions commence at very early embryonic stages and coordination remains crucial for survival. The underlying molecular mechanisms of pacemaker cell development and function are still not fully understood. Heart form and function show high evolutionary conservation. Even in simple contractile cardiac tubes in primitive invertebrates, cardiac function is controlled by intrinsic, autonomous pacemaker cells. Understanding the evolutionary origin and development of cardiac pacemaker cells will help us outline the important pathways and factors involved. Key patterning factors, such as the homeodomain transcription factors Nkx2.5 and Shox2, and the LIM-homeodomain transcription factor Islet-1, components of the T-box (Tbx), and bone morphogenic protein (Bmp) families are well conserved. Here we compare the dominant pacemaking systems in various organisms with respect to the underlying molecular regulation. Comparative analysis of the pathways involved in patterning the pacemaker domain in an evolutionary context might help us outline a common fundamental pacemaker cell gene programme. Special focus is given to pacemaker development in zebrafish, an extensively used model for vertebrate development. Finally, we conclude with a summary of highly conserved key factors in pacemaker cell development and function.
Collapse
Affiliation(s)
- Silja Burkhard
- Hubrecht Institute-KNAW and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands.
| | - Vincent van Eif
- Department of Medical Biology, Academic Medical Center Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Laurence Garric
- Hubrecht Institute-KNAW and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands.
| | - Vincent M Christoffels
- Department of Medical Biology, Academic Medical Center Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands.
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands.
| |
Collapse
|
41
|
|
42
|
Galinsky R, Lear CA, Yamaguchi K, Wassink G, Westgate JA, Bennet L, Gunn AJ. Cholinergic and β-adrenergic control of cardiovascular reflex responses to brief repeated asphyxia in term-equivalent fetal sheep. Am J Physiol Regul Integr Comp Physiol 2016; 311:R949-R956. [PMID: 27654399 DOI: 10.1152/ajpregu.00340.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/12/2016] [Indexed: 11/22/2022]
Abstract
The role of cholinergic and β-adrenergic activity in mediating fetal cardiovascular recovery from brief repeated episodes of asphyxia consistent with established labor, remains unclear. In this study, we tested the effect of cholinergic and β-adrenergic blockade on the fetal chemoreflex and fetal heart rate (FHR) overshoot responses during brief repeated asphyxia at rates consistent with early or active labor. Chronically instrumented fetal sheep at 0.85 of gestation received either i.v. atropine sulfate (cholinergic blockade, n=8) or vehicle (n=7) followed by 3 x 1-minute umbilical cord occlusions repeated every 5 minutes (1:5; consistent with early labor), or i.v. propranolol hydrochloride (β-adrenergic blockade, n=6) or vehicle (n=6) followed by 3 x 2-minute occlusions repeated every 5 minutes (2:5; consistent with active labor). In vehicle-controls, 1:5 occlusions were associated with rapid and sustained FHR decelerations followed by rapid return of FHR to baseline values after release of the occlusion. Cholinergic blockade abolished FHR decelerations during occlusions and caused FHR overshoot after release of the occlusion (P<0.05 vs. control 1:5). In vehicle-controls, 2:5 occlusions caused rapid and sustained FHR decelerations followed by FHR overshoot after release of the occlusion. β-adrenergic blockade was associated with greater reduction in FHR during occlusions and attenuated FHR overshoot (P<0.05 vs. control 2:5). These data demonstrate that the FHR overshoot pattern after asphyxia is mediated by a combination of attenuated parasympathetic activity and increased β-adrenergic stimulation of the fetal heart.
Collapse
|