1
|
Duan Z, Cao H, Xu M, Huang W, Peng Y, Shen Z, Hu S, Han Y. Chemogenetic ablation and regeneration of arterial valve in zebrafish. Biochem Biophys Res Commun 2025; 762:151786. [PMID: 40209504 DOI: 10.1016/j.bbrc.2025.151786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 03/28/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Aortic valve diseases are prevalent and severe cardiovascular conditions with limited treatment options beyond surgical intervention. The ability to regenerate aortic valves would revolutionize the management of these diseases. Utilizing the zebrafish model, which possesses remarkable regenerative capacities, we developed a chemogenetic arterial valve ablation model using a zebrafish-codon optimized nitroreductase. We found that arterial valve ablation led to blood regurgitation and impaired cardiac function, which are commonly associated with aortic valve diseases. Following ablation, zebrafish arterial valve could fully regenerate and restore valvular and cardiac function. Moreover, suppression of blood flow significantly impedes valve regeneration, indicating the importance of hemodynamic forces in this process. Our research has successfully established a robust aortic valve injury model to study the cellular and molecular mechanisms underlying its regeneration process which will facilitate the development of innovative therapeutic strategies tailored for aortic valve diseases.
Collapse
Affiliation(s)
- Zongyi Duan
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Hao Cao
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Mengting Xu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Wenping Huang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Yuanhui Peng
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Zhenya Shen
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Shijun Hu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Yanchao Han
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China.
| |
Collapse
|
2
|
Derrick CJ, Eley L, Alqahtani A, Henderson DJ, Chaudhry B. Zebrafish arterial valve development occurs through direct differentiation of second heart field progenitors. Cardiovasc Res 2025; 121:157-173. [PMID: 39460530 PMCID: PMC11998914 DOI: 10.1093/cvr/cvae230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/03/2024] [Accepted: 09/15/2024] [Indexed: 10/28/2024] Open
Abstract
AIMS Bicuspid aortic valve (BAV) is the most common congenital heart defect, affecting at least 2% of the population. The embryonic origins of BAV remain poorly understood, with few assays for validating patient variants, limiting the identification of causative genes for BAV. In both human and mouse, the left and right leaflets of the arterial valves arise from the outflow tract cushions, with interstitial cells originating from neural crest cells and the overlying endocardium through endothelial-to-mesenchymal transition (EndoMT). In contrast, an EndoMT-independent mechanism of direct differentiation of cardiac progenitors from the second heart field (SHF) is responsible for the formation of the anterior and posterior leaflets. Defects in either of these developmental mechanisms can result in BAV. Although zebrafish have been suggested as a model for human variant testing, their naturally bicuspid arterial valve has not been considered suitable for understanding human arterial valve development. Here, we have set out to investigate to what extent the processes involved in arterial valve development are conserved in zebrafish and, ultimately, whether functional testing of BAV variants could be carried out. METHODS AND RESULTS Using a combination of live imaging, immunohistochemistry, and Cre-mediated lineage tracing, we show that the zebrafish arterial valve primordia develop directly from SHF progenitors with no contribution from EndoMT or neural crest, in keeping with the human and mouse anterior and posterior leaflets. Moreover, once formed, these primordia share common subsequent developmental events with all three aortic valve leaflets. CONCLUSION Our work highlights a conserved ancestral mechanism of arterial valve leaflet formation from the SHF and identifies that development of the arterial valve is distinct from that of the atrioventricular valve in zebrafish. Crucially, this confirms the utility of zebrafish for understanding the development of specific BAV subtypes and arterial valve dysplasia, offering potential for high-throughput variant testing.
Collapse
Affiliation(s)
- Christopher J Derrick
- International Centre for Life, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Lorraine Eley
- International Centre for Life, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Ahlam Alqahtani
- International Centre for Life, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Deborah J Henderson
- International Centre for Life, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Bill Chaudhry
- International Centre for Life, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| |
Collapse
|
3
|
Ji X, Zou Y, Guan W, Su X, Yuan J, Li Q, Lu Z, Xiao J, Huang H, Wang M, Guo Z. Comparative analysis of the heart histological structure, metabolic enzyme activities and transcriptome profiles of juvenile and adult yellowfin tuna (Thunnus albacares) in the South China Sea. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101460. [PMID: 40037069 DOI: 10.1016/j.cbd.2025.101460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
The yellowfin tuna is a large marine carnivorous fish with high commercial value. It is known for its unique physiological characteristics and holds significant potential for aquaculture. However, research on this species' developmental biology and physiology remains limited, particularly regarding the structural characteristics and functional changes in the developing heart. To investigate the differences in cardiac tissue structure and function at different developmental stages in yellowfin tuna, we conducted comparative analyses of histology, metabolic enzyme activity, and transcriptomes. Hematoxylin and eosin (H&E) and Masson staining revealed that cardiac muscle fibers were thicker and more compact, and the area of collagen fibers was significantly increased in adult fish compared to juvenile fish (p < 0.001). Additionally, the enzyme activities of Na+K+-ATPase, Ca2+Mg2+-ATPase, carnitine palmitoyltransferase 1 (CPT-1), lactate dehydrogenase (LDH), succinate dehydrogenase (SDH), and malate dehydrogenase (MDH) were notably greater in adult fish compared to juvenile fish (p < 0.05). Comparative transcriptome analysis identified 1293 differentially expressed genes (DEGs) between juvenile and adult fish. Functional enrichment analyses indicated that these differential genes are primarily closely associated with heart development and metabolic regulation pathways. Furthermore, key metabolism-related DEGs, such as acsl3b, acsbg2, acsl1a, and cpt1ab, were further identified, and quantitative real-time PCR (qRT-PCR) validated the accuracy of the results. In conclusion, this study provides a systematic analysis of the differences in histology, metabolic enzyme activities, and transcriptomics between the hearts of juvenile and adult yellowfin tuna, providing foundational data for future research on heart development in the later stages of yellowfin tuna and contributing to the advancement of aquaculture practices for this species.
Collapse
Affiliation(s)
- Xu Ji
- School of Life and Health Sciences, State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou 570228, China
| | - Ying Zou
- School of Life and Health Sciences, State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou 570228, China
| | - Wanlin Guan
- School of Life and Health Sciences, State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou 570228, China
| | - Xiameng Su
- School of Life and Health Sciences, State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou 570228, China
| | - Jigui Yuan
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Qian Li
- School of Life and Health Sciences, State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou 570228, China
| | - Zhiyuan Lu
- College of Marine Science and Engineering, Hainan University, Haikou 570228, China
| | - Juan Xiao
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Hai Huang
- Key Laboratory of Utilization and Conservation for Tropical Marine Bioresources, Hainan Key Laboratory for Conservation and Utilization of Tropical Marine Fishery Resources, College of Fisheries and Life Science, Hainan Tropical Ocean University, Sanya 572022, China
| | - Mei Wang
- College of Marine Science and Engineering, Hainan University, Haikou 570228, China.
| | - Zhiqiang Guo
- School of Life and Health Sciences, State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou 570228, China; College of Marine Science and Engineering, Hainan University, Haikou 570228, China.
| |
Collapse
|
4
|
Sánchez-Posada J, Derrick CJ, Noël ES. morphoHeart: A quantitative tool for integrated 3D morphometric analyses of heart and ECM during embryonic development. PLoS Biol 2025; 23:e3002995. [PMID: 39879226 PMCID: PMC11778784 DOI: 10.1371/journal.pbio.3002995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025] Open
Abstract
Heart development involves the complex structural remodelling of a linear heart tube into an asymmetrically looped and ballooned organ. Previous studies have associated regional expansion of extracellular matrix (ECM) space with tissue morphogenesis during development. We have developed morphoHeart, a 3D tissue segmentation and morphometry software with a user-friendly graphical interface (GUI) that delivers the first integrated 3D visualisation and multiparametric analysis of both heart and ECM morphology in live embryos. morphoHeart reveals that the ECM undergoes regional dynamic expansion and reduction during cardiac development, concomitant with chamber-specific morphological maturation. We use morphoHeart to demonstrate that regionalised ECM expansion driven by the ECM crosslinker Hapln1a promotes atrial lumen expansion during heart development. Finally, morphoHeart's GUI expands its use beyond that of cardiac tissue, allowing its segmentation and morphometric analysis tools to be applied to z-stack images of any fluorescently labelled tissue.
Collapse
Affiliation(s)
- Juliana Sánchez-Posada
- School of Biosciences and Bateson Centre, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Christopher J. Derrick
- Biosciences Institute, Faculty of Biomedical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Emily S. Noël
- School of Biosciences and Bateson Centre, University of Sheffield, Western Bank, Sheffield, United Kingdom
| |
Collapse
|
5
|
López-Unzu MA, Teresa Soto-Navarrete M, Sans-Coma V, Fernández B, Carmen Durán A. The myoarchitecture of the vertebrate cardiac ventricles: evolution and classification. J Exp Biol 2024; 227:jeb247441. [PMID: 39392075 DOI: 10.1242/jeb.247441] [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] [Indexed: 10/12/2024]
Abstract
The ventricle of the vertebrate heart is the main segment of the cardiac outflow region. Compared with other cardiac components, it shows remarkable histomorphological variation among different animal groups. This variation is especially apparent in the myocardium, which is generally classified into three main types: trabeculated, compact and mixed. The trabeculated or 'spongy' myocardium is characterized by the existence of trabeculae and deep recesses or intertrabecular spaces, lined by the endocardium. The compact type is composed of condensed myocardial fibers, with almost no trabeculated layer. The mixed type consists of an outer compact layer and an inner trabeculated layer. Among vertebrates, fishes show a great diversity of myocardial types. On this basis, the ventricular myoarchitecture has been categorized into four groups of varying complexity. This classification is made according to (i) the proportion of the two types of myocardium, trabeculated versus compact, and (ii) the vascularization of the heart wall. Here, we review the morphogenetic mechanisms that give rise to the different ventricular myoarchitecture in gnathostomes (i.e. jawed vertebrates) with special emphasis on the diversity of the ventricular myocardium throughout the phylogeny of ancient actinopterygians and teleosts. Finally, we propose that the classification of the ventricular myoarchitecture should be reconsidered, given that the degrees of myocardial compactness on which the current classification system is based do not constitute discrete states, but an anatomical continuum.
Collapse
Affiliation(s)
- Miguel A López-Unzu
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - María Teresa Soto-Navarrete
- Departamento de Biología Animal, Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29590 Málaga, Spain
| | - Valentín Sans-Coma
- Departamento de Biología Animal, Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29590 Málaga, Spain
| | - Borja Fernández
- Departamento de Biología Animal, Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29590 Málaga, Spain
- Instituto de Biotecnología y Desarrollo Azul-IBYDA, 29004 Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares-CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ana Carmen Durán
- Departamento de Biología Animal, Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29590 Málaga, Spain
- Instituto de Biotecnología y Desarrollo Azul-IBYDA, 29004 Málaga, Spain
| |
Collapse
|
6
|
Goossens E, Deblock L, Caboor L, Eynden DVD, Josipovic I, Isaacura PR, Maksimova E, Van Impe M, Bonnin A, Segers P, Cornillie P, Boone MN, Van Driessche I, De Spiegelaere W, De Roo J, Sips P, De Buysser K. From Corrosion Casting to Virtual Dissection: Contrast-Enhanced Vascular Imaging using Hafnium Oxide Nanocrystals. SMALL METHODS 2024; 8:e2301499. [PMID: 38200600 DOI: 10.1002/smtd.202301499] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Indexed: 01/12/2024]
Abstract
Vascular corrosion casting is a method used to visualize the three dimensional (3D) anatomy and branching pattern of blood vessels. A polymer resin is injected in the vascular system and, after curing, the surrounding tissue is removed. The latter often deforms or even fractures the fragile cast. Here, a method is proposed that does not require corrosion, and is based on in situ micro computed tomography (micro-CT) scans. To overcome the lack of CT contrast between the polymer cast and the animals' surrounding soft tissue, hafnium oxide nanocrystals (HfO2 NCs) are introduced as CT contrast agents into the resin. The NCs dramatically improve the overall CT contrast of the cast and allow for straightforward segmentation in the CT scans. Careful design of the NC surface chemistry ensures the colloidal stability of the NCs in the casting resin. Using only 5 m% of HfO2 NCs, high-quality cardiovascular casts of both zebrafish and mice can be automatically segmented using CT imaging software. This allows to differentiate even μ $\umu$ m-scale details without having to alter the current resin injection methods. This new method of virtual dissection by visualizing casts in situ using contrast-enhanced CT imaging greatly expands the application potential of the technique.
Collapse
Affiliation(s)
- Eline Goossens
- Department of Chemistry, Ghent University, Ghent, 9000, Belgium
- Department of Chemistry, University of Basel, Basel, 4058, Switzerland
| | - Loren Deblock
- Department of Chemistry, Ghent University, Ghent, 9000, Belgium
| | - Lisa Caboor
- Department of Biomolecular Medicine, Ghent University, Ghent, 9000, Belgium
| | - Dietger Van den Eynden
- Department of Chemistry, Ghent University, Ghent, 9000, Belgium
- Department of Chemistry, University of Basel, Basel, 4058, Switzerland
| | - Iván Josipovic
- Center for X-ray Tomography, Ghent University, Ghent, 9000, Belgium
| | - Pablo Reyes Isaacura
- Laboratory of Veterinary Morphology, Ghent University, Merelbeke, 9820, Belgium
- Centre for Polymer Material Technologies, Ghent University, Ghent, 9052, Belgium
- Laboratory for Chemical Technology, Ghent University, Ghent, 9052, Belgium
| | - Elizaveta Maksimova
- Department of Chemistry, University of Basel, Basel, 4058, Switzerland
- Swiss Light Source, Paul Scherrer Institut, Villigen PSI, 5232, Switzerland
- Swiss Nanoscience Institute, University of Basel, Basel, 4056, Switzerland
| | - Matthias Van Impe
- Institute of Biomedical Engineering and Technology, Ghent University, Ghent, 9000, Belgium
| | - Anne Bonnin
- Swiss Light Source, Paul Scherrer Institut, Villigen PSI, 5232, Switzerland
| | - Patrick Segers
- Institute of Biomedical Engineering and Technology, Ghent University, Ghent, 9000, Belgium
| | - Pieter Cornillie
- Laboratory of Veterinary Morphology, Ghent University, Merelbeke, 9820, Belgium
| | - Matthieu N Boone
- Center for X-ray Tomography, Ghent University, Ghent, 9000, Belgium
| | | | - Ward De Spiegelaere
- Laboratory of Veterinary Morphology, Ghent University, Merelbeke, 9820, Belgium
| | - Jonathan De Roo
- Department of Chemistry, University of Basel, Basel, 4058, Switzerland
| | - Patrick Sips
- Department of Biomolecular Medicine, Ghent University, Ghent, 9000, Belgium
| | | |
Collapse
|
7
|
Chakraborty S, Bhattacharya S, Meyers BA, Sepúlveda MS, Vlachos PP. Evolution of cardiac tissue and flow mechanics in developing Japanese Medaka. PLoS One 2024; 19:e0309018. [PMID: 39186731 PMCID: PMC11346936 DOI: 10.1371/journal.pone.0309018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 08/03/2024] [Indexed: 08/28/2024] Open
Abstract
The effects of pressure drop across cardiac valve cushion regions and endocardial wall strain in the early developmental stages of a teleost species heart are poorly understood. In the presented work, we utilize microscale particle image velocimetry (μPIV) flow measurements of developing medaka hearts from 3 to 14 dpf (n = 5 at each dpf) to quantify the pressure field and endocardial wall strain. Peak pressure drop at the atrioventricular canal (ΔPAVC) and outflow tract (ΔPOFT) show a steady increase with fish age progression. Pressure drops when non-dimensionalized with blood viscosity and heart rate at each dpf are comparable with measurements in zebrafish hearts. Retrograde flows captured at these regions display a negative pressure drop. A novel metric, Endocardial Work (EW), is introduced by analyzing the ΔPAVC-strain curves, which is a non-invasive measure of work required for ventricle filling. EW is a metric that can differentiate between the linear heart stage (< 100 Pa-%), cardiac looped chamber stage (< 300 Pa-%), and the fully formed chamber stage (> 300 Pa-%).
Collapse
Affiliation(s)
- Sreyashi Chakraborty
- Department of Mechanical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| | - Sayantan Bhattacharya
- Department of Mechanical Engineering, University of Maryland, Baltimore County, Baltimore, Maryland, United States of America
| | - Brett Albert Meyers
- Department of Mechanical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| | - Maria S. Sepúlveda
- Department of Forestry & Natural Resources, Purdue University, West Lafayette, Indiana, United States of America
| | - Pavlos P. Vlachos
- Department of Mechanical Engineering, Purdue University, West Lafayette, Indiana, United States of America
- Department of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| |
Collapse
|
8
|
Angom RS, Joshi A, Patowary A, Sivadas A, Ramasamy S, K. V. S, Kaushik K, Sabharwal A, Lalwani MK, K. S, Singh N, Scaria V, Sivasubbu S. Forward genetic screen using a gene-breaking trap approach identifies a novel role of grin2bb-associated RNA transcript ( grin2bbART) in zebrafish heart function. Front Cell Dev Biol 2024; 12:1339292. [PMID: 38533084 PMCID: PMC10964321 DOI: 10.3389/fcell.2024.1339292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/23/2024] [Indexed: 03/28/2024] Open
Abstract
LncRNA-based control affects cardiac pathophysiologies like myocardial infarction, coronary artery disease, hypertrophy, and myotonic muscular dystrophy. This study used a gene-break transposon (GBT) to screen zebrafish (Danio rerio) for insertional mutagenesis. We identified three insertional mutants where the GBT captured a cardiac gene. One of the adult viable GBT mutants had bradycardia (heart arrhythmia) and enlarged cardiac chambers or hypertrophy; we named it "bigheart." Bigheart mutant insertion maps to grin2bb or N-methyl D-aspartate receptor (NMDAR2B) gene intron 2 in reverse orientation. Rapid amplification of adjacent cDNA ends analysis suggested a new insertion site transcript in the intron 2 of grin2bb. Analysis of the RNA sequencing of wild-type zebrafish heart chambers revealed a possible new transcript at the insertion site. As this putative lncRNA transcript satisfies the canonical signatures, we called this transcript grin2bb associated RNA transcript (grin2bbART). Using in situ hybridization, we confirmed localized grin2bbART expression in the heart, central nervous system, and muscles in the developing embryos and wild-type adult zebrafish atrium and bulbus arteriosus. The bigheart mutant had reduced Grin2bbART expression. We showed that bigheart gene trap insertion excision reversed cardiac-specific arrhythmia and atrial hypertrophy and restored grin2bbART expression. Morpholino-mediated antisense downregulation of grin2bbART in wild-type zebrafish embryos mimicked bigheart mutants; this suggests grin2bbART is linked to bigheart. Cardiovascular tissues use Grin2bb as a calcium-permeable ion channel. Calcium imaging experiments performed on bigheart mutants indicated calcium mishandling in the heart. The bigheart cardiac transcriptome showed differential expression of calcium homeostasis, cardiac remodeling, and contraction genes. Western blot analysis highlighted Camk2d1 and Hdac1 overexpression. We propose that altered calcium activity due to disruption of grin2bbART, a putative lncRNA in bigheart, altered the Camk2d-Hdac pathway, causing heart arrhythmia and hypertrophy in zebrafish.
Collapse
Affiliation(s)
- Ramcharan Singh Angom
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL, United States
| | - Adita Joshi
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Ashok Patowary
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Ambily Sivadas
- GN Ramachandran Knowledge Center for Genome Informatics, Council of Scientific and Industrial Research, Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Soundhar Ramasamy
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Shamsudheen K. V.
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
- GN Ramachandran Knowledge Center for Genome Informatics, Council of Scientific and Industrial Research, Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Kriti Kaushik
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Ankit Sabharwal
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Mukesh Kumar Lalwani
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Subburaj K.
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Naresh Singh
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Vinod Scaria
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
- GN Ramachandran Knowledge Center for Genome Informatics, Council of Scientific and Industrial Research, Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Sridhar Sivasubbu
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| |
Collapse
|
9
|
Schindler M, Endlich N. Zebrafish as a model for podocyte research. Am J Physiol Renal Physiol 2024; 326:F369-F381. [PMID: 38205541 DOI: 10.1152/ajprenal.00335.2023] [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: 10/18/2023] [Revised: 12/25/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Podocytes, specialized postmitotic cells, are central players in various kidney-related diseases. Zebrafish have become a valuable model system for studying podocyte biology because they are genetically easy to manipulate, transparent, and their glomerular structure is similar to that of mammals. This review provides an overview of the knowledge of podocyte biology in zebrafish larvae, with particular focus on their essential contribution to understanding the mechanisms that underlie kidney diseases as well as supporting drug development. In addition, special attention is given to advances in live-imaging techniques allowing the observation of dynamic processes, including podocyte motility, podocyte process behavior, and glomerulus maturation. The review further addresses the functional aspects of podocytes in zebrafish larvae. This includes topics such as glomerular filtration, ultrastructural analyses, and evaluation of podocyte response to nephrotoxic insults. Studies presented in this context have provided important insights into the maintenance and resistance of the glomerular filtration barrier in zebrafish larvae and explored the potential transferability of these findings to mammals such as mice, rats, and most importantly, humans. The recent ability to identify potential therapeutic targets represents a promising new way to identify drugs that could effectively treat podocyte-associated glomerulopathies in humans. In summary, this review gives an overview about the importance of zebrafish as a model for podocyte-related disease and targeted drug development. It also highlights the key role of advanced imaging techniques in transparent zebrafish larvae, improving our understanding of glomerular diseases and the significant potential for translation of these findings to humans.
Collapse
Affiliation(s)
- Maximilian Schindler
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
10
|
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
|
11
|
Rolland L, Abaroa JM, Faucherre A, Drouard A, Jopling C. The ion channel Trpc6a regulates the cardiomyocyte regenerative response to mechanical stretch. Front Cardiovasc Med 2024; 10:1186086. [PMID: 38259319 PMCID: PMC10801195 DOI: 10.3389/fcvm.2023.1186086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/26/2023] [Indexed: 01/24/2024] Open
Abstract
Myocardial damage caused, for example, by cardiac ischemia leads to ventricular volume overload resulting in increased stretch of the remaining myocardium. In adult mammals, these changes trigger an adaptive cardiomyocyte hypertrophic response which, if the damage is extensive, will ultimately lead to pathological hypertrophy and heart failure. Conversely, in response to extensive myocardial damage, cardiomyocytes in the adult zebrafish heart and neonatal mice proliferate and completely regenerate the damaged myocardium. We therefore hypothesized that in adult zebrafish, changes in mechanical loading due to myocardial damage may act as a trigger to induce cardiac regeneration. Based on this notion we sought to identify mechanosensors which could be involved in detecting changes in mechanical loading and triggering regeneration. Here we show using a combination of knockout animals, RNAseq and in vitro assays that the mechanosensitive ion channel Trpc6a is required by cardiomyocytes for successful cardiac regeneration in adult zebrafish. Furthermore, using a cyclic cell stretch assay, we have determined that Trpc6a induces the expression of components of the AP1 transcription complex in response to mechanical stretch. Our data highlights how changes in mechanical forces due to myocardial damage can be detected by mechanosensors which in turn can trigger cardiac regeneration.
Collapse
Affiliation(s)
| | | | | | | | - Chris Jopling
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, LabEx ICST, Montpellier, France
| |
Collapse
|
12
|
Sukumaran V, Mutlu O, Murtaza M, Alhalbouni R, Dubansky B, Yalcin HC. Experimental assessment of cardiovascular physiology in the chick embryo. Dev Dyn 2023; 252:1247-1268. [PMID: 37002896 DOI: 10.1002/dvdy.589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 12/13/2022] [Accepted: 03/10/2023] [Indexed: 10/04/2023] Open
Abstract
High resolution assessment of cardiac functional parameters is crucial in translational animal research. The chick embryo is a historically well-used in vivo model for cardiovascular research due to its many practical advantages, and the conserved form and function of the chick and human cardiogenesis programs. This review aims to provide an overview of several different technical approaches for chick embryo cardiac assessment. Doppler echocardiography, optical coherence tomography, micromagnetic resonance imaging, microparticle image velocimetry, real-time pressure monitoring, and associated issues with the techniques will be discussed. Alongside this discussion, we also highlight recent advances in cardiac function measurements in chick embryos.
Collapse
Affiliation(s)
| | - Onur Mutlu
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | | | - Benjamin Dubansky
- Department of Biological and Agricultural Engineering, Office of Research and Economic Development, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Huseyin C Yalcin
- Biomedical Research Center, Qatar University, Doha, Qatar
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
13
|
Shaftoe JB, Manchester EA, Gillis TE. Cardiac remodeling caused by cold acclimation is reversible with rewarming in zebrafish (Danio rerio). Comp Biochem Physiol A Mol Integr Physiol 2023; 283:111466. [PMID: 37302568 DOI: 10.1016/j.cbpa.2023.111466] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/31/2023] [Accepted: 06/08/2023] [Indexed: 06/13/2023]
Abstract
Cold acclimation of zebrafish causes changes to the structure and composition of the heart. However, little is known of the consequences of these changes on heart function or if these changes are reversible with rewarming back to the initial temperature. In the current study, zebrafish were acclimated from 27℃ to 20°C, then after 17 weeks, a subset of fish were rewarmed to 27°C and held at that temperature for 7 weeks. The length of this trial, 23 weeks, was chosen to mimic seasonal changes in temperature. Cardiac function was measured in each group at 27°C and 20°C using high frequency ultrasound. It was found that cold acclimation caused a decrease in ventricular cross-sectional area, compact myocardial thickness, and total muscle area. There was also a decrease in end-diastolic area with cold acclimation that reversed upon rewarming to control temperatures. Rewarming caused an increase in the thickness of the compact myocardium, total muscle area, and end-diastolic area back to control levels. This is the first experiment to demonstrate that cardiac remodeling, induced by cold acclimation, is reversible upon re-acclimation to control temperature (27°C). Finally, body condition measurements reveal that fish that had been cold-acclimated and then reacclimated to 27°C, were in poorer condition than the fish that remained at 20°C as well as the control fish at week 23. This suggests that the physiological responses to the multiple changes in temperature had a significant energetic cost to the animal. SUMMARY STATEMENT: The decrease in cardiac muscle density, compact myocardium thickness and diastolic area in zebrafish caused by cold acclimation, was reversed with rewarming to control temperatures.
Collapse
Affiliation(s)
- Jared B Shaftoe
- Department of Integrative Biology, University of Guelph, Canada
| | | | - Todd E Gillis
- Department of Integrative Biology, University of Guelph, Canada.
| |
Collapse
|
14
|
Mizukami K, Higashiyama H, Arima Y, Ando K, Okada N, Kose K, Yamada S, Takeuchi JK, Koshiba-Takeuchi K, Fukuhara S, Miyagawa-Tomita S, Kurihara H. Coronary artery established through amniote evolution. eLife 2023; 12:e83005. [PMID: 37605519 PMCID: PMC10444023 DOI: 10.7554/elife.83005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 07/17/2023] [Indexed: 08/23/2023] Open
Abstract
Coronary arteries are a critical part of the vascular system and provide nourishment to the heart. In humans, even minor defects in coronary arteries can be lethal, emphasizing their importance for survival. However, some teleosts survive without coronary arteries, suggesting that there may have been some evolutionary changes in the morphology and function of coronary arteries in the tetrapod lineage. Here, we propose that the true ventricular coronary arteries were newly established during amniote evolution through remodeling of the ancestral coronary vasculature. In mouse (Mus musculus) and Japanese quail (Coturnix japonica) embryos, the coronary arteries unique to amniotes are established by the reconstitution of transient vascular plexuses: aortic subepicardial vessels (ASVs) in the outflow tract and the primitive coronary plexus on the ventricle. In contrast, amphibians (Hyla japonica, Lithobates catesbeianus, Xenopus laevis, and Cynops pyrrhogaster) retain the ASV-like vasculature as truncal coronary arteries throughout their lives and have no primitive coronary plexus. The anatomy and development of zebrafish (Danio rerio) and chondrichthyans suggest that their hypobranchial arteries are ASV-like structures serving as the root of the coronary vasculature throughout their lives. Thus, the ventricular coronary artery of adult amniotes is a novel structure that has acquired a new remodeling process, while the ASVs, which occur transiently during embryonic development, are remnants of the ancestral coronary vessels. This evolutionary change may be related to the modification of branchial arteries, indicating considerable morphological changes underlying the physiological transition during amniote evolution.
Collapse
Affiliation(s)
- Kaoru Mizukami
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of TokyoTokyoJapan
| | - Hiroki Higashiyama
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of TokyoTokyoJapan
| | - Yuichiro Arima
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of TokyoTokyoJapan
- Developmental Cardiology Laboratory, International Research Center for Medical Science, Kumamoto UniversityKumamotoJapan
| | - Koji Ando
- Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical SchoolTokyoJapan
| | | | - Katsumi Kose
- Institute of Applied Physics, University of TsukubaTsukubaJapan
| | - Shigehito Yamada
- Congenital Anomaly Research Center, Kyoto University Graduate School of MedicineKyotoJapan
| | - Jun K Takeuchi
- Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental UniversityTokyoJapan
| | | | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical SchoolTokyoJapan
| | - Sachiko Miyagawa-Tomita
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of TokyoTokyoJapan
- Heart Center, Department of Pediatric Cardiology, Tokyo Women’s Medical UniversityTokyoJapan
- Department of Animal Nursing Science, Yamazaki University of Animal Health TechnologyTokyoJapan
| | - Hiroki Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of TokyoTokyoJapan
| |
Collapse
|
15
|
Dalla Barba F, Soardi M, Mouhib L, Risato G, Akyürek EE, Lucon-Xiccato T, Scano M, Benetollo A, Sacchetto R, Richard I, Argenton F, Bertolucci C, Carotti M, Sandonà D. Modeling Sarcoglycanopathy in Danio rerio. Int J Mol Sci 2023; 24:12707. [PMID: 37628888 PMCID: PMC10454440 DOI: 10.3390/ijms241612707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Sarcoglycanopathies, also known as limb girdle muscular dystrophy 3-6, are rare muscular dystrophies characterized, although heterogeneous, by high disability, with patients often wheelchair-bound by late adolescence and frequently developing respiratory and cardiac problems. These diseases are currently incurable, emphasizing the importance of effective treatment strategies and the necessity of animal models for drug screening and therapeutic verification. Using the CRISPR/Cas9 genome editing technique, we generated and characterized δ-sarcoglycan and β-sarcoglycan knockout zebrafish lines, which presented a progressive disease phenotype that worsened from a mild larval stage to distinct myopathic features in adulthood. By subjecting the knockout larvae to a viscous swimming medium, we were able to anticipate disease onset. The δ-SG knockout line was further exploited to demonstrate that a δ-SG missense mutant is a substrate for endoplasmic reticulum-associated degradation (ERAD), indicating premature degradation due to protein folding defects. In conclusion, our study underscores the utility of zebrafish in modeling sarcoglycanopathies through either gene knockout or future knock-in techniques. These novel zebrafish lines will not only enhance our understanding of the disease's pathogenic mechanisms, but will also serve as powerful tools for phenotype-based drug screening, ultimately contributing to the development of a cure for sarcoglycanopathies.
Collapse
Affiliation(s)
- Francesco Dalla Barba
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy; (F.D.B.)
| | - Michela Soardi
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy; (F.D.B.)
| | - Leila Mouhib
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy; (F.D.B.)
- Randall Center for Cell and Molecular Biophysics, King’s College London, London WC2R 2LS, UK
| | - Giovanni Risato
- Department of Biology, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Via Giustiniani, 2, 35128 Padova, Italy
| | - Eylem Emek Akyürek
- Department of Comparative Biomedicine and Food Science, University of Padova, Agripolis, Legnaro, 35020 Padova, Italy
| | - Tyrone Lucon-Xiccato
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Martina Scano
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy; (F.D.B.)
| | - Alberto Benetollo
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy; (F.D.B.)
| | - Roberta Sacchetto
- Department of Comparative Biomedicine and Food Science, University of Padova, Agripolis, Legnaro, 35020 Padova, Italy
| | - Isabelle Richard
- Genethon, F-91002 Evry, France
- INSERM, U951, INTEGRARE Research Unit, F-91002 Evry, France
| | - Francesco Argenton
- Department of Biology, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy
| | - Cristiano Bertolucci
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Marcello Carotti
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy; (F.D.B.)
| | - Dorianna Sandonà
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy; (F.D.B.)
| |
Collapse
|
16
|
Zhang J, Zhu J, Xie T, Sun F, Wang N, Guo FM. Quantitative evaluation of myocardial layer-specific strain using two-dimensional speckle tracking echocardiography in septic patients. BMC Anesthesiol 2023; 23:271. [PMID: 37568093 PMCID: PMC10416394 DOI: 10.1186/s12871-023-02186-x] [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: 03/27/2023] [Accepted: 06/18/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Although global longitudinal strain (GLS) is proven to be reduced and associated with adverse outcomes in septic patients, it has not been elucidated whether or not layer-specific strains are reduced. We aimed to explore the layer-specific strains of left ventricular (LV) for assessing myocardial dysfunction in septic patients. METHODS A prospective observational study of patients with sepsis was conducted in a tertiary hospital in China. Routine two-dimensional speckle tracking echocardiography was performed within 24 h of enrollment. Demographic data, laboratory values, and clinical outcomes were collected. RESULTS We recruited 79 septic patients finally. The mean age of septic patients was 59.4 years old and 45 (57.0%) were male. The median Acute Physiology Age and Chronic Health Evaluation (APACHE II) score, and mean sequential organ failure assessment (SOFA) score of all patients were 19.0 and 7.7, respectively. According to the left ventricular ejection fraction (LVEF) value of 50%, the patients were categorized into two groups: SICM (sepsis-induced cardiomyopathy, LVEF < 50%, n = 22) and non-SICM group ( LVEF ≥ 50%, n = 57). The median LVEF of SICM and non-SICM patients were 41.9% and 58.7%, and SICM patients had less negative layer-specific strain and global strain than that of non-SICM patients. The echocardiographic comparison of non-SICM and healthy controls was conducted to explore the myocardial injuries of non-SICM patients and the non-SICM had worse LS-epi than that of controls (-18.5% vs. -21.4%, p = 0.024). CONCLUSION There were 72.2% (57) septic patients presented with non-SICM (LVEF ≥ 50%), and the strain value of epicardium of them was less negative than healthy controls.
Collapse
Affiliation(s)
- Jin Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jing Zhu
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Tian Xie
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Fan Sun
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Ni Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Feng-Mei Guo
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
17
|
Sree Kumar H, Wisner AS, Refsnider JM, Martyniuk CJ, Zubcevic J. Small fish, big discoveries: zebrafish shed light on microbial biomarkers for neuro-immune-cardiovascular health. Front Physiol 2023; 14:1186645. [PMID: 37324381 PMCID: PMC10267477 DOI: 10.3389/fphys.2023.1186645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Zebrafish (Danio rerio) have emerged as a powerful model to study the gut microbiome in the context of human conditions, including hypertension, cardiovascular disease, neurological disorders, and immune dysfunction. Here, we highlight zebrafish as a tool to bridge the gap in knowledge in linking the gut microbiome and physiological homeostasis of cardiovascular, neural, and immune systems, both independently and as an integrated axis. Drawing on zebrafish studies to date, we discuss challenges in microbiota transplant techniques and gnotobiotic husbandry practices. We present advantages and current limitations in zebrafish microbiome research and discuss the use of zebrafish in identification of microbial enterotypes in health and disease. We also highlight the versatility of zebrafish studies to further explore the function of human conditions relevant to gut dysbiosis and reveal novel therapeutic targets.
Collapse
Affiliation(s)
- Hemaa Sree Kumar
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH, United States
- Department of Neuroscience and Neurological Disorders, University of Toledo, Toledo, OH, United States
| | - Alexander S. Wisner
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, OH, United States
- Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, United States
| | - Jeanine M. Refsnider
- Department of Environmental Sciences, University of Toledo, Toledo, OH, United States
| | - Christopher J. Martyniuk
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, OH, United States
| | - Jasenka Zubcevic
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH, United States
| |
Collapse
|
18
|
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
|
19
|
Coppola A, Lombari P, Mazzella E, Capolongo G, Simeoni M, Perna AF, Ingrosso D, Borriello M. Zebrafish as a Model of Cardiac Pathology and Toxicity: Spotlight on Uremic Toxins. Int J Mol Sci 2023; 24:ijms24065656. [PMID: 36982730 PMCID: PMC10052014 DOI: 10.3390/ijms24065656] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
Chronic kidney disease (CKD) is an increasing health care problem. About 10% of the general population is affected by CKD, representing the sixth cause of death in the world. Cardiovascular events are the main mortality cause in CKD, with a cardiovascular risk 10 times higher in these patients than the rate observed in healthy subjects. The gradual decline of the kidney leads to the accumulation of uremic solutes with a negative effect on every organ, especially on the cardiovascular system. Mammalian models, sharing structural and functional similarities with humans, have been widely used to study cardiovascular disease mechanisms and test new therapies, but many of them are rather expensive and difficult to manipulate. Over the last few decades, zebrafish has become a powerful non-mammalian model to study alterations associated with human disease. The high conservation of gene function, low cost, small size, rapid growth, and easiness of genetic manipulation are just some of the features of this experimental model. More specifically, embryonic cardiac development and physiological responses to exposure to numerous toxin substances are similar to those observed in mammals, making zebrafish an ideal model to study cardiac development, toxicity, and cardiovascular disease.
Collapse
Affiliation(s)
- Annapaola Coppola
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Patrizia Lombari
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Elvira Mazzella
- Department of Translational Medical Science, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Giovanna Capolongo
- Department of Translational Medical Science, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Mariadelina Simeoni
- Department of Translational Medical Science, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Alessandra F. Perna
- Department of Translational Medical Science, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Diego Ingrosso
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Margherita Borriello
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Correspondence:
| |
Collapse
|
20
|
Ontogenetic changes in the body structure of the Arctic fish Leptoclinus maculatus. Sci Rep 2023; 13:3688. [PMID: 36879005 PMCID: PMC9988964 DOI: 10.1038/s41598-023-30251-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Histological studies of the ontogenetic changes in Arctic marine fishes are often fragmented and incomplete. Here we present a comprehensive histological ontogenetic analysis of the daubed shanny (Leptoclinus maculatus) from the Arctic, characterizing its development as it undergoes a series of changes in the organ and tissue organization, especially during the postlarvae transition from the pelagic to benthic lifestyle. The thyroid, heart, digestive tract, liver, gonads, blood, and the lipid sac of the postlarvae at different developmental stages (L1-L5) were studied for the first time. We found that L. maculatus has structural characteristics of marine fish developing in cold, high-oxygen polar waters. We conclude that the presence of the lipid sac and the absence of distinguishable red blood cells in pelagic postlarvae are unique features of the daubed shanny most likely linked to its successful growth and development in the Arctic environment.
Collapse
|
21
|
Salehin N, Teranikar T, Lee J, Chuong CJ. Ventricular anisotropic deformation and contractile function of the developing heart of zebrafish in vivo. Dev Dyn 2023; 252:247-262. [PMID: 36057940 DOI: 10.1002/dvdy.536] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 08/10/2022] [Accepted: 08/29/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND The developing zebrafish ventricle generates higher intraventricular pressure (IVP) with increasing stroke volume and cardiac output at different developmental stages to meet the metabolic demands of the rapidly growing embryo (Salehin et al. Ann Biomed Eng, 2021;49(9): 2080-2093). To understand the changing role of the developing embryonic heart, we studied its biomechanical characteristics during in vivo cardiac cycles. By combining changes in wall strains and IVP measurements, we assessed ventricular wall stiffness during diastolic filling and the ensuing systolic IVP-generation capacity during 3-, 4-, and 5-day post fertilization (dpf). We further examined the anisotropy of wall deformation, in different regions within the ventricle, throughout a complete cardiac cycle. RESULTS We found the ventricular walls grow increasingly stiff during diastolic filling with a corresponding increase in IVP-generation capacity from 3- to 4- and 5-dpf groups. In addition, we found the corresponding increasing level of anisotropic wall deformation through cardiac cycles that favor the latitudinal direction, with the most pronounced found in the equatorial region of the ventricle. CONCLUSIONS From 3- to 4- and 5-dpf groups, the ventricular wall myocardium undergoes increasing level of anisotropic deformation. This, in combination with the increasing wall stiffness and IVP-generation capacity, allows the developing heart to effectively pump blood to meet the rapidly growing embryo's needs.
Collapse
Affiliation(s)
- Nabid Salehin
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Tanveer Teranikar
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Juhyun Lee
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Cheng-Jen Chuong
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| |
Collapse
|
22
|
Akerberg AA, Trembley M, Butty V, Schwertner A, Zhao L, Beerens M, Liu X, Mahamdeh M, Yuan S, Boyer L, MacRae C, Nguyen C, Pu WT, Burns CE, Burns CG. RBPMS2 Is a Myocardial-Enriched Splicing Regulator Required for Cardiac Function. Circ Res 2022; 131:980-1000. [PMID: 36367103 PMCID: PMC9770155 DOI: 10.1161/circresaha.122.321728] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND RBPs (RNA-binding proteins) perform indispensable functions in the post-transcriptional regulation of gene expression. Numerous RBPs have been implicated in cardiac development or physiology based on gene knockout studies and the identification of pathogenic RBP gene mutations in monogenic heart disorders. The discovery and characterization of additional RBPs performing indispensable functions in the heart will advance basic and translational cardiovascular research. METHODS We performed a differential expression screen in zebrafish embryos to identify genes enriched in nkx2.5-positive cardiomyocytes or cardiopharyngeal progenitors compared to nkx2.5-negative cells from the same embryos. We investigated the myocardial-enriched gene RNA-binding protein with multiple splicing (variants) 2 [RBPMS2)] by generating and characterizing rbpms2 knockout zebrafish and human cardiomyocytes derived from RBPMS2-deficient induced pluripotent stem cells. RESULTS We identified 1848 genes enriched in the nkx2.5-positive population. Among the most highly enriched genes, most with well-established functions in the heart, we discovered the ohnologs rbpms2a and rbpms2b, which encode an evolutionarily conserved RBP. Rbpms2 localizes selectively to cardiomyocytes during zebrafish heart development and strong cardiomyocyte expression persists into adulthood. Rbpms2-deficient embryos suffer from early cardiac dysfunction characterized by reduced ejection fraction. The functional deficit is accompanied by myofibril disarray, altered calcium handling, and differential alternative splicing events in mutant cardiomyocytes. These phenotypes are also observed in RBPMS2-deficient human cardiomyocytes, indicative of conserved molecular and cellular function. RNA-sequencing and comparative analysis of genes mis-spliced in RBPMS2-deficient zebrafish and human cardiomyocytes uncovered a conserved network of 29 ortholog pairs that require RBPMS2 for alternative splicing regulation, including RBFOX2, SLC8A1, and MYBPC3. CONCLUSIONS Our study identifies RBPMS2 as a conserved regulator of alternative splicing, myofibrillar organization, and calcium handling in zebrafish and human cardiomyocytes.
Collapse
Affiliation(s)
- Alexander A. Akerberg
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Michael Trembley
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Vincent Butty
- BioMicroCenter, Department of Biology (V.B.), Massachusetts Institute of Technology, Cambridge‚ MA
- Department of Biology (V.B., L.B.), Massachusetts Institute of Technology, Cambridge‚ MA
| | - Asya Schwertner
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Long Zhao
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Manu Beerens
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA (M.B., C.M.)
| | - Xujie Liu
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Mohammed Mahamdeh
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Shiaulou Yuan
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Laurie Boyer
- Department of Biology (V.B., L.B.), Massachusetts Institute of Technology, Cambridge‚ MA
- Department of Biological Engineering (L.B.), Massachusetts Institute of Technology, Cambridge‚ MA
| | - Calum MacRae
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA (M.B., C.M.)
| | - Christopher Nguyen
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
- Cardiovascular Innovation Research Center, Heart Vascular & Thoracic Institute, Cleveland Clinic‚ Cleveland‚ OH (C.N.)
| | - William T. Pu
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
- Harvard Stem Cell Institute, Cambridge, MA (W.T.P., C.E.B.)
| | - Caroline E. Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
- Harvard Stem Cell Institute, Cambridge, MA (W.T.P., C.E.B.)
| | - C. Geoffrey Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| |
Collapse
|
23
|
Birkedal R, Laasmaa M, Branovets J, Vendelin M. Ontogeny of cardiomyocytes: ultrastructure optimization to meet the demand for tight communication in excitation-contraction coupling and energy transfer. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210321. [PMID: 36189816 PMCID: PMC9527910 DOI: 10.1098/rstb.2021.0321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The ontogeny of the heart describes its development from the fetal to the adult stage. In newborn mammals, blood pressure and thus cardiac performance are relatively low. The cardiomyocytes are thin, and with a central core of mitochondria surrounded by a ring of myofilaments, while the sarcoplasmic reticulum (SR) is sparse. During development, as blood pressure and performance increase, the cardiomyocytes become more packed with structures involved in excitation–contraction (e-c) coupling (SR and myofilaments) and the generation of ATP (mitochondria) to fuel the contraction. In parallel, the e-c coupling relies increasingly on calcium fluxes through the SR, while metabolism relies increasingly on fatty acid oxidation. The development of transverse tubules and SR brings channels and transporters interacting via calcium closer to each other and is crucial for e-c coupling. However, for energy transfer, it may seem counterintuitive that the increased structural density restricts the overall ATP/ADP diffusion. In this review, we discuss how this is because of the organization of all these structures forming modules. Although the overall diffusion across modules is more restricted, the energy transfer within modules is fast. A few studies suggest that in failing hearts this modular design is disrupted, and this may compromise intracellular energy transfer. This article is part of the theme issue ‘The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease’.
Collapse
Affiliation(s)
- Rikke Birkedal
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Akadeemia 15, room SCI-218, 12618 Tallinn, Estonia
| | - Martin Laasmaa
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Akadeemia 15, room SCI-218, 12618 Tallinn, Estonia
| | - Jelena Branovets
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Akadeemia 15, room SCI-218, 12618 Tallinn, Estonia
| | - Marko Vendelin
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Akadeemia 15, room SCI-218, 12618 Tallinn, Estonia
| |
Collapse
|
24
|
Mackrill JJ. Evolution of the cardiac dyad. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210329. [PMID: 36189805 PMCID: PMC9527923 DOI: 10.1098/rstb.2021.0329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/22/2021] [Indexed: 12/30/2022] Open
Abstract
Cardiac dyads are the site of communication between the sarcoplasmic reticulum (SR) and infoldings of the sarcolemma called transverse-tubules (TT). During heart excitation-contraction coupling, Ca2+-influx through L-type Ca2+ channels in the TT is amplified by release of Ca2+-from the SR via type 2 ryanodine receptors, activating the contractile apparatus. Key proteins involved in cardiac dyad function are bridging integrator 1 (BIN1), junctophilin 2 and caveolin 3. The work presented here aims to reconstruct the evolutionary history of the cardiac dyad, by surveying the scientific literature for ultrastructural evidence of these junctions across all animal taxa; phylogenetically reconstructing the evolutionary history of BIN1; and by comparing peptide motifs involved in TT formation by this protein across metazoans. Key findings are that cardiac dyads have been identified in mammals, arthropods and molluscs, but not in other animals. Vertebrate BIN1 does not group with members of this protein family from other taxa, suggesting that invertebrate BINs are paralogues rather orthologues of this gene. Comparisons of BIN1 peptide sequences of mammals with those of other vertebrates reveals novel features that might contribute to TT and dyad formation. The analyses presented here suggest that the cardiac dyad evolved independently several times during metazoan evolution: an unexpected observation given the diversity of heart structure and function between different animal taxa. This article is part of the theme issue 'The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease'.
Collapse
Affiliation(s)
- John James Mackrill
- Department of Physiology, School of Medicine, University College Cork, Western Gateway Building, Western Road, Cork T12 XF62, Republic of Ireland
| |
Collapse
|
25
|
Sun J, Peterson EA, Jiao C, Chen X, Zhao Y, Wang J. Zebrafish heart regeneration after coronary dysfunction-induced cardiac damage. Dev Biol 2022; 487:57-66. [PMID: 35490764 PMCID: PMC11017783 DOI: 10.1016/j.ydbio.2022.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 11/03/2022]
Abstract
Over the past 20 years, various zebrafish injury models demonstrated efficient heart regeneration after cardiac tissue loss. However, no established coronary vessel injury methods exist in the zebrafish model, despite coronary endothelial dysfunction occurring in most patients with acute coronary syndrome. This is due to difficulties performing surgery on small coronary vessels and a lack of genetic tools to precisely manipulate coronary cells in zebrafish. We determined that the Notch ligand gene deltaC regulatory sequences drive gene expression in zebrafish coronary endothelial cells, enabling us to overcome these obstacles. We created a deltaC fluorescent reporter line and visualized robust coronary growth during heart development and regeneration. Importantly, this reporter facilitated the visualization of coronary growth without an endocardial background. Moreover, we visualized robust coronary growth on the surface of juvenile hearts and regrowth in the wounded area of adult hearts ex vivo. With this approach, we observed growth inhibition by reported vascular growth antagonists of the VEGF, EGF and Notch signaling pathways. Furthermore, we established a coronary genetic ablation system and observed that severe coronary endothelial cell loss resulted in fish death, whereas fish survived mild coronary cell loss. Coronary cell depletion triggered regenerative responses, which resulted in the restoration of damaged cardiac tissues within several weeks. Overall, our work demonstrated the efficacy of using deltaC regulatory elements for high-resolution visualization of the coronary endothelium; screening small molecules for coronary growth effects; and revealed complete recovery in adult zebrafish after coronary-induced heart damage.
Collapse
Affiliation(s)
- Jisheng Sun
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Elizabeth A Peterson
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Cheng Jiao
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Xin Chen
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Yun Zhao
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Jinhu Wang
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
26
|
Abstract
Heart disease is the leading cause of death worldwide. Despite decades of research, most heart pathologies have limited treatments, and often the only curative approach is heart transplantation. Thus, there is an urgent need to develop new therapeutic approaches for treating cardiac diseases. Animal models that reproduce the human pathophysiology are essential to uncovering the biology of diseases and discovering therapies. Traditionally, mammals have been used as models of cardiac disease, but the cost of generating and maintaining new models is exorbitant, and the studies have very low throughput. In the last decade, the zebrafish has emerged as a tractable model for cardiac diseases, owing to several characteristics that made this animal popular among developmental biologists. Zebrafish fertilization and development are external; embryos can be obtained in high numbers, are cheap and easy to maintain, and can be manipulated to create new genetic models. Moreover, zebrafish exhibit an exceptional ability to regenerate their heart after injury. This review summarizes 25 years of research using the zebrafish to study the heart, from the classical forward screenings to the contemporary methods to model mutations found in patients with cardiac disease. We discuss the advantages and limitations of this model organism and introduce the experimental approaches exploited in zebrafish, including forward and reverse genetics and chemical screenings. Last, we review the models used to induce cardiac injury and essential ideas derived from studying natural regeneration. Studies using zebrafish have the potential to accelerate the discovery of new strategies to treat cardiac diseases.
Collapse
Affiliation(s)
- Juan Manuel González-Rosa
- Cardiovascular Research Center, Massachusetts General Hospital Research Institute, Harvard Medical School, Charlestown, MA
| |
Collapse
|
27
|
Abrial M, Basu S, Huang M, Butty V, Schwertner A, Jeffrey S, Jordan D, Burns CE, Burns CG. Latent TGFβ-binding proteins 1 and 3 protect the larval zebrafish outflow tract from aneurysmal dilatation. Dis Model Mech 2022; 15:dmm046979. [PMID: 35098309 PMCID: PMC8990920 DOI: 10.1242/dmm.046979] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 01/13/2022] [Indexed: 11/20/2022] Open
Abstract
Aortic root aneurysm is a common cause of morbidity and mortality in Loeys-Dietz and Marfan syndromes, where perturbations in transforming growth factor beta (TGFβ) signaling play a causal or contributory role, respectively. Despite the advantages of cross-species disease modeling, animal models of aortic root aneurysm are largely restricted to genetically engineered mice. Here, we report that zebrafish devoid of the genes encoding latent-transforming growth factor beta-binding protein 1 and 3 (ltbp1 and ltbp3, respectively) develop rapid and severe aneurysm of the outflow tract (OFT), the aortic root equivalent. Similar to syndromic aneurysm tissue, the distended OFTs display evidence for paradoxical hyperactivated TGFβ signaling. RNA-sequencing revealed significant overlap between the molecular signatures of disease tissue from mutant zebrafish and a mouse model of Marfan syndrome. Moreover, chemical inhibition of TGFβ signaling in wild-type animals phenocopied mutants but chemical activation did not, demonstrating that TGFβ signaling is protective against aneurysm. Human relevance is supported by recent studies implicating genetic lesions in LTBP3 and, potentially, LTBP1 as heritable causes of aortic root aneurysm. Ultimately, our data demonstrate that zebrafish can now be leveraged to interrogate thoracic aneurysmal disease and identify novel lead compounds through small-molecule suppressor screens. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Maryline Abrial
- Cardiovascular Research Center, Department of Cardiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Sandeep Basu
- Harvard Medical School, Boston, MA 02115, USA
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Mengmeng Huang
- Cardiovascular Research Center, Department of Cardiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Vincent Butty
- BioMicroCenter, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Asya Schwertner
- Cardiovascular Research Center, Department of Cardiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Spencer Jeffrey
- Cardiovascular Research Center, Department of Cardiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Daniel Jordan
- Cardiovascular Research Center, Department of Cardiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Caroline E. Burns
- Cardiovascular Research Center, Department of Cardiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - C. Geoffrey Burns
- Cardiovascular Research Center, Department of Cardiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| |
Collapse
|
28
|
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
|
29
|
Hoareau M, El Kholti N, Debret R, Lambert E. Zebrafish as a Model to Study Vascular Elastic Fibers and Associated Pathologies. Int J Mol Sci 2022; 23:2102. [PMID: 35216218 PMCID: PMC8875079 DOI: 10.3390/ijms23042102] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/09/2022] [Accepted: 02/12/2022] [Indexed: 02/06/2023] Open
Abstract
Many extensible tissues such as skin, lungs, and blood vessels require elasticity to function properly. The recoil of elastic energy stored during a stretching phase is provided by elastic fibers, which are mostly composed of elastin and fibrillin-rich microfibrils. In arteries, the lack of elastic fibers leads to a weakening of the vessel wall with an increased risk to develop cardiovascular defects such as stenosis, aneurysms, and dissections. The development of new therapeutic molecules involves preliminary tests in animal models that recapitulate the disease and whose response to drugs should be as close as possible to that of humans. Due to its superior in vivo imaging possibilities and the broad tool kit for forward and reverse genetics, the zebrafish has become an important model organism to study human pathologies. Moreover, it is particularly adapted to large scale studies, making it an attractive model in particular for the first steps of investigations. In this review, we discuss the relevance of the zebrafish model for the study of elastic fiber-related vascular pathologies. We evidence zebrafish as a compelling alternative to conventional mouse models.
Collapse
Affiliation(s)
- Marie Hoareau
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Institut de Biologie et Chimie des Protéines, Université Lyon 1, 7, Passage du Vercors, CEDEX 07, F-69367 Lyon, France; (N.E.K.); (R.D.)
| | | | | | - Elise Lambert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Institut de Biologie et Chimie des Protéines, Université Lyon 1, 7, Passage du Vercors, CEDEX 07, F-69367 Lyon, France; (N.E.K.); (R.D.)
| |
Collapse
|
30
|
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
|
31
|
Rosa A, Giese W, Meier K, Alt S, Klaus-Bergmann A, Edgar LT, Bartels E, Collins R, Szymborska A, Coxam B, Bernabeu MO, Gerhardt H. Wasp controls oriented migration of endothelial cells to achieve functional vascular patterning. Development 2021; 149:273808. [PMID: 34931661 PMCID: PMC8918813 DOI: 10.1242/dev.200195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/10/2021] [Indexed: 11/21/2022]
Abstract
Endothelial cell migration and proliferation are essential for the establishment of a hierarchical organization of blood vessels and optimal distribution of blood. However, how these cellular processes are quantitatively coordinated to drive vascular network morphogenesis remains unknown. Here, using the zebrafish vasculature as a model system, we demonstrate that the balanced distribution of endothelial cells, as well as the resulting regularity of vessel calibre, is a result of cell migration from veins towards arteries and cell proliferation in veins. We identify the Wiskott-Aldrich Syndrome protein (WASp) as an important molecular regulator of this process and show that loss of coordinated migration from veins to arteries upon wasb depletion results in aberrant vessel morphology and the formation of persistent arteriovenous shunts. We demonstrate that WASp achieves its function through the coordination of junctional actin assembly and PECAM1 recruitment and provide evidence that this is conserved in humans. Overall, we demonstrate that functional vascular patterning in the zebrafish trunk is established through differential cell migration regulated by junctional actin, and that interruption of differential migration may represent a pathomechanism in vascular malformations. Summary: Regular diameter of developing veins and arteries in the zebrafish trunk is controlled by differential endothelial cell proliferation and WASp-driven directed cell migration.
Collapse
Affiliation(s)
- André Rosa
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Wolfgang Giese
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Katja Meier
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Silvanus Alt
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Alexandra Klaus-Bergmann
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Lowell T Edgar
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Eireen Bartels
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Russell Collins
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Anna Szymborska
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Baptiste Coxam
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Miguel O Bernabeu
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK.,The Bayes Centre, The University of Edinburgh, Edinburgh, United Kingdom. 5 Berlin Institute of Health (BIH), Berlin, Germany
| | - Holger Gerhardt
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| |
Collapse
|
32
|
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
|
33
|
Ando K, Shih YH, Ebarasi L, Grosse A, Portman D, Chiba A, Mattonet K, Gerri C, Stainier DYR, Mochizuki N, Fukuhara S, Betsholtz C, Lawson ND. Conserved and context-dependent roles for pdgfrb signaling during zebrafish vascular mural cell development. Dev Biol 2021; 479:11-22. [PMID: 34310924 DOI: 10.1016/j.ydbio.2021.06.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 06/17/2021] [Indexed: 12/27/2022]
Abstract
Platelet derived growth factor beta and its receptor, Pdgfrb, play essential roles in the development of vascular mural cells, including pericytes and vascular smooth muscle cells. To determine if this role was conserved in zebrafish, we analyzed pdgfb and pdgfrb mutant lines. Similar to mouse, pdgfb and pdgfrb mutant zebrafish lack brain pericytes and exhibit anatomically selective loss of vascular smooth muscle coverage. Despite these defects, pdgfrb mutant zebrafish did not otherwise exhibit circulatory defects at larval stages. However, beginning at juvenile stages, we observed severe cranial hemorrhage and vessel dilation associated with loss of pericytes and vascular smooth muscle cells in pdgfrb mutants. Similar to mouse, pdgfrb mutant zebrafish also displayed structural defects in the glomerulus, but normal development of hepatic stellate cells. We also noted defective mural cell investment on coronary vessels with concomitant defects in their development. Together, our studies support a conserved requirement for Pdgfrb signaling in mural cells. In addition, these zebrafish mutants provide an important model for definitive investigation of mural cells during early embryonic stages without confounding secondary effects from circulatory defects.
Collapse
Affiliation(s)
- Koji Ando
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds Väg 20, SE-751 85, Uppsala, Sweden; Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School, Sendagi Bunkyo-ku, Tokyo, 113 8602, Japan.
| | - Yu-Huan Shih
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01650, United States
| | - Lwaki Ebarasi
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden
| | - Ann Grosse
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01650, United States
| | - Daneal Portman
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01650, United States
| | - Ayano Chiba
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, 564 8565, Japan
| | - Kenny Mattonet
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231, Bad Nauheim, Germany
| | - Claudia Gerri
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231, Bad Nauheim, Germany; Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231, Bad Nauheim, Germany
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, 564 8565, Japan
| | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School, Sendagi Bunkyo-ku, Tokyo, 113 8602, Japan
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds Väg 20, SE-751 85, Uppsala, Sweden; Department of Medicine Huddinge (MedH), Karolinska Institutet, Campus Flemingsberg, Neo, Blickagången 16, Hiss S, Plan 7, SE-141 57, Huddinge, Sweden
| | - Nathan D Lawson
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01650, United States.
| |
Collapse
|
34
|
Zhao Y, Chen C, Yun M, Issa T, Lin A, Nguyen TP. Constructing Adult Zebrafish Einthoven's Triangle to Define Electrical Heart Axes. Front Physiol 2021; 12:708938. [PMID: 34366897 PMCID: PMC8342992 DOI: 10.3389/fphys.2021.708938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/24/2021] [Indexed: 11/19/2022] Open
Abstract
Zebrafish is a popular high-throughput vertebrate model to study human cardiac electrophysiology, arrhythmias, and myopathies. One reason for this popularity is the purported striking similarities between zebrafish and human electrocardiograms (ECGs). However, zebrafish electrical heart axes were unknown. It is impossible to define heart axis based on single-lead ECG because determination of an electrical heart axis in the frontal plane requires the use of the hexaxial reference system (or Cabrera system) derived from Einthoven’s triangle. Construction of Einthoven’s triangle requires simultaneous ECG recording from at least two Einthoven bipolar leads. Therefore, we systematically constructed the first zebrafish Einthoven’s triangle by simultaneous bipolar dual-lead ECG recording to determine for the first time the three frontal electrical heart axes using the Cabrera system. Comparing zebrafish with human Einthoven’s triangle reveals that their normal frontal electrical axes were reflections of each other across 0° in the Cabrera system. The responsible mechanisms involve zebrafish vs. human cardiac activation propagating in the same direction along the heart horizontal axis but in opposite directions along the heart longitudinal axis. The same observations are true for zebrafish vs. human cardiac repolarization. This study marks a technical breakthrough in the first bipolar dual-lead ECG recording in live adult zebrafish to construct for the first time zebrafish Einthoven’s triangle. This first systematic analysis of the actual differences and similarities between normal adult zebrafish and human Einthoven’s triangles unmasked differences and similarities in the underlying cardiac axis mechanisms. Insights of the live adult zebrafish main heart axis and its three frontal electrical heart axes provide critical contextual framework to interpret the clinical relevance of the adult zebrafish heart as model for human cardiac electrophysiology.
Collapse
Affiliation(s)
- Yali Zhao
- The Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Connie Chen
- The Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Morgan Yun
- The Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Thomas Issa
- The Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Andrew Lin
- The Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Thao P Nguyen
- The Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
35
|
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
|
36
|
Kirillova A, Han L, Liu H, Kühn B. Polyploid cardiomyocytes: implications for heart regeneration. Development 2021; 148:271050. [PMID: 34897388 DOI: 10.1242/dev.199401] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Terminally differentiated cells are generally thought to have arrived at their final form and function. Many terminally differentiated cell types are polyploid, i.e. they have multiple copies of the normally diploid genome. Mammalian heart muscle cells, termed cardiomyocytes, are one such example of polyploid cells. Terminally differentiated cardiomyocytes are bi- or multi-nucleated, or have polyploid nuclei. Recent mechanistic studies of polyploid cardiomyocytes indicate that they can limit cellular proliferation and, hence, heart regeneration. In this short Spotlight, we present the mechanisms generating bi- and multi-nucleated cardiomyocytes, and the mechanisms generating polyploid nuclei. Our aim is to develop hypotheses about how these mechanisms might relate to cardiomyocyte proliferation and cardiac regeneration. We also discuss how these new findings could be applied to advance cardiac regeneration research, and how they relate to studies of other polyploid cells, such as cancer cells.
Collapse
Affiliation(s)
- Anna Kirillova
- Medical Scientist Training Program, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA 15219, USA
| | - Lu Han
- Division of Cardiology, UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA.,Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA
| | - Honghai Liu
- Division of Cardiology, UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA.,Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA
| | - Bernhard Kühn
- Division of Cardiology, UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA.,Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA.,McGowan Institute of Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| |
Collapse
|
37
|
Hodges MM, Zgheib C, Liechty KW. A Large Mammalian Model of Myocardial Regeneration After Myocardial Infarction in Fetal Sheep. Adv Wound Care (New Rochelle) 2021; 10:174-190. [PMID: 32496979 DOI: 10.1089/wound.2018.0894] [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] [Indexed: 11/13/2022] Open
Abstract
Objective: Ischemic heart disease accounts for over 20% of all deaths worldwide. As the global population faces a rising burden of chronic diseases, such as hypertension, hyperlipidemia, and diabetes, the prevalence of heart failure due to ischemic heart disease is estimated to increase. We sought to develop a model that may more accurately identify therapeutic targets to mitigate the development of heart failure following myocardial infarction (MI). Approach: Having utilized fetal large mammalian models of scarless wound healing, we proposed a fetal ovine model of myocardial regeneration after MI. Results: Use of this model has identified critical pathways in the mammalian response to MI, which are differentially activated in the regenerative, fetal mammalian response to MI when compared to the reparative, scar-forming, adult mammalian response to MI. Innovation: While the foundation of myocardial regeneration research has been built on zebrafish and rodent models, effective therapies derived from these disease models have been lacking; therefore, we sought to develop a more representative ovine model of myocardial regeneration after MI to improve the identification of therapeutic targets designed to mitigate the development of heart failure following MI. Conclusions: To develop therapies aimed at mitigating this rising burden of disease, it is critical that the animal models we utilize closely reflect the physiology and pathology we observe in human disease. We encourage use of this ovine large mammalian model to facilitate identification of therapies designed to mitigate the growing burden of heart failure.
Collapse
Affiliation(s)
- Maggie M. Hodges
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Carlos Zgheib
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Kenneth W. Liechty
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, Colorado, USA
| |
Collapse
|
38
|
Computational Modeling of Blood Flow Hemodynamics for Biomechanical Investigation of Cardiac Development and Disease. J Cardiovasc Dev Dis 2021; 8:jcdd8020014. [PMID: 33572675 PMCID: PMC7912127 DOI: 10.3390/jcdd8020014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/16/2021] [Accepted: 01/21/2021] [Indexed: 12/11/2022] Open
Abstract
The heart is the first functional organ in a developing embryo. Cardiac development continues throughout developmental stages while the heart goes through a serious of drastic morphological changes. Previous animal experiments as well as clinical observations showed that disturbed hemodynamics interfere with the development of the heart and leads to the formation of a variety of defects in heart valves, heart chambers, and blood vessels, suggesting that hemodynamics is a governing factor for cardiogenesis, and disturbed hemodynamics is an important source of congenital heart defects. Therefore, there is an interest to image and quantify the flowing blood through a developing heart. Flow measurement in embryonic fetal heart can be performed using advanced techniques such as magnetic resonance imaging (MRI) or echocardiography. Computational fluid dynamics (CFD) modeling is another approach especially useful when the other imaging modalities are not available and in-depth flow assessment is needed. The approach is based on numerically solving relevant physical equations to approximate the flow hemodynamics and tissue behavior. This approach is becoming widely adapted to simulate cardiac flows during the embryonic development. While there are few studies for human fetal cardiac flows, many groups used zebrafish and chicken embryos as useful models for elucidating normal and diseased cardiogenesis. In this paper, we explain the major steps to generate CFD models for simulating cardiac hemodynamics in vivo and summarize the latest findings on chicken and zebrafish embryos as well as human fetal hearts.
Collapse
|
39
|
Helston O, Amaya E. Reactive oxygen species during heart regeneration in zebrafish: Lessons for future clinical therapies. Wound Repair Regen 2021; 29:211-224. [PMID: 33471940 PMCID: PMC8611801 DOI: 10.1111/wrr.12892] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/30/2020] [Accepted: 12/22/2020] [Indexed: 11/30/2022]
Abstract
In humans, myocardial infarction (MI) is associated with irreversible damage to heart tissue, resulting in increased morbidity and mortality in patients. By comparison, the zebrafish (Danio rerio) is capable of repairing damaged and injured hearts by activating a full regenerative response. By studying model organisms that can regenerate loss heart tissue following injury, such as the zebrafish, a greater insight will be gained into the molecular pathways that can induce and sustain a regenerative response following injury. There is hope that such information may lead to new treatments or therapies aimed at stimulating a better regenerative response in humans that have suffered heart attacks. Recent findings in zebrafish have highlighted an important role for sustained elevated levels of Reactive Oxygen Species (ROS), including hydrogen peroxide (H2O2) in the promotion of a regenerative response. Given that elevated levels of H2O2 can be harmful, simply elevating ROS levels directly may not be easy or practical to translate clinically. An alternative approach would be to identify the critical downstream targets of ROS in the promotion of heart regeneration, and then target these clinically using drugs. One such family of potential downstream targets of ROS during heart regeneration are the family of protein tyrosine phosphatases (PTPs), which are known to be exquisitely sensitive to redox regulation and whose inhibition have been linked to the promotion of heart regeneration in zebrafish. In this review, we present an overview of the zebrafish as a model organism for studying cardiac regeneration, including the molecular mechanisms by which cardiac regeneration occurs in response to injury. We then present recent findings linking elevated ROS levels to heart regeneration and their potential downstream targets, the PTPs, including protein tyrosine phosphatase 1B (PTP1B) and the dual specificity phosphatase 6 (DUSP6) in the promotion of heart regeneration.
Collapse
Affiliation(s)
- Olivia Helston
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Enrique Amaya
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
40
|
Bazmi M, Escobar AL. Excitation-Contraction Coupling in the Goldfish ( Carassius auratus) Intact Heart. Front Physiol 2020; 11:1103. [PMID: 33041845 PMCID: PMC7518121 DOI: 10.3389/fphys.2020.01103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/10/2020] [Indexed: 12/17/2022] Open
Abstract
Cardiac physiology of fish models is an emerging field given the ease of genome editing and the development of transgenic models. Several studies have described the cardiac properties of zebrafish (Denio rerio). The goldfish (Carassius auratus) belongs to the same family as the zebrafish and has emerged as an alternative model with which to study cardiac function. Here, we propose to acutely study electrophysiological and systolic Ca2+ signaling in intact goldfish hearts. We assessed the Ca2+ dynamics and the electrophysiological cardiac function of goldfish, zebrafish, and mice models, using pulsed local field fluorescence microscopy, intracellular microelectrodes, and flash photolysis in perfused hearts. We observed goldfish ventricular action potentials (APs) and Ca2+ transients to be significantly longer when compared to the zebrafish. The action potential half duration at 50% (APD50) of goldfish was 370.38 ± 8.8 ms long, and in the zebrafish they were observed to be only 83.9 ± 9.4 ms. Additionally, the half duration of the Ca2+ transients was also longer for goldfish (402.1 ± 4.4 ms) compared to the zebrafish (99.1 ± 2.7 ms). Also, blocking of the L-type Ca2+ channels with nifedipine revealed this current has a major role in defining the amplitude and the duration of goldfish Ca2+ transients. Interestingly, nifedipine flash photolysis experiments in the intact heart identified whether or not the decrease in the amplitude of Ca2+ transients was due to shorter APs. Moreover, an increase in temperature and heart rate had a strong shortening effect on the AP and Ca2+ transients of goldfish hearts. Furthermore, ryanodine (Ry) and thapsigargin (Tg) significantly reduced the amplitude of the Ca2+ transients, induced a prolongation in the APs, and altogether exhibited the degree to which the Ca2+ release from the sarcoplasmic reticulum contributed to the Ca2+ transients. We conclude that the electrophysiological properties and Ca2+ signaling in intact goldfish hearts strongly resembles the endocardial layer of larger mammals.
Collapse
Affiliation(s)
- Maedeh Bazmi
- Quantitative Systems Biology Program, School of Natural Sciences, University of California, Merced, Merced, CA, United States
| | - Ariel L Escobar
- Department of Bioengineering, School of Engineering, University of California, Merced, Merced, CA, United States
| |
Collapse
|
41
|
Michel JB. Phylogenic Determinants of Cardiovascular Frailty, Focus on Hemodynamics and Arterial Smooth Muscle Cells. Physiol Rev 2020; 100:1779-1837. [DOI: 10.1152/physrev.00022.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The evolution of the circulatory system from invertebrates to mammals has involved the passage from an open system to a closed in-parallel system via a closed in-series system, accompanying the increasing complexity and efficiency of life’s biological functions. The archaic heart enables pulsatile motion waves of hemolymph in invertebrates, and the in-series circulation in fish occurs with only an endothelium, whereas mural smooth muscle cells appear later. The present review focuses on evolution of the circulatory system. In particular, we address how and why this evolution took place from a closed, flowing, longitudinal conductance at low pressure to a flowing, highly pressurized and bifurcating arterial compartment. However, although arterial pressure was the latest acquired hemodynamic variable, the general teleonomy of the evolution of species is the differentiation of individual organ function, supported by specific fueling allowing and favoring partial metabolic autonomy. This was achieved via the establishment of an active contractile tone in resistance arteries, which permitted the regulation of blood supply to specific organ activities via its localized function-dependent inhibition (active vasodilation). The global resistance to viscous blood flow is the peripheral increase in frictional forces caused by the tonic change in arterial and arteriolar radius, which backscatter as systemic arterial blood pressure. Consequently, the arterial pressure gradient from circulating blood to the adventitial interstitium generates the unidirectional outward radial advective conductance of plasma solutes across the wall of conductance arteries. This hemodynamic evolution was accompanied by important changes in arterial wall structure, supported by smooth muscle cell functional plasticity, including contractility, matrix synthesis and proliferation, endocytosis and phagocytosis, etc. These adaptive phenotypic shifts are due to epigenetic regulation, mainly related to mechanotransduction. These paradigms actively participate in cardio-arterial pathologies such as atheroma, valve disease, heart failure, aneurysms, hypertension, and physiological aging.
Collapse
|
42
|
Gendernalik A, Zebhi B, Ahuja N, Garrity D, Bark D. In Vivo Pressurization of the Zebrafish Embryonic Heart as a Tool to Characterize Tissue Properties During Development. Ann Biomed Eng 2020; 49:834-845. [PMID: 32959136 DOI: 10.1007/s10439-020-02619-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/10/2020] [Indexed: 12/31/2022]
Abstract
Cardiac morphogenesis requires an intricate orchestration of mechanical stress to sculpt the heart as it transitions from a straight tube to a multichambered adult heart. Mechanical properties are fundamental to this process, involved in a complex interplay with function, morphology, and mechanotransduction. In the current work, we propose a pressurization technique applied to the zebrafish atrium to quantify mechanical properties of the myocardium under passive tension. By further measuring deformation, we obtain a pressure-stretch relationship that is used to identify constitutive models of the zebrafish embryonic cardiac tissue. Two-dimensional results are compared with a three-dimensional finite element analysis based on reconstructed embryonic heart geometry. Through these steps, we found that the myocardium of zebrafish results in a stiffness on the order of 10 kPa immediately after the looping stage of development. This work enables the ability to determine how these properties change under normal and pathological heart development.
Collapse
Affiliation(s)
- Alex Gendernalik
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Banafsheh Zebhi
- Department of Mechanical Engineering, Colorado State University, Room 304 Scott Building, 1374 Campus Delivery, Fort Collins, CO, 80523-1374, USA
| | - Neha Ahuja
- Department of Biology, Colorado State University, Fort Collins, CO, USA
| | - Deborah Garrity
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA.,Department of Biology, Colorado State University, Fort Collins, CO, USA
| | - David Bark
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA. .,Department of Mechanical Engineering, Colorado State University, Room 304 Scott Building, 1374 Campus Delivery, Fort Collins, CO, 80523-1374, USA. .,Department of Pediatrics, University of Colorado, Aurora, CO, USA. .,Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
43
|
Abstract
The valves of the heart are crucial for ensuring that blood flows in one direction from the heart, through the lungs and back to the rest of the body. Heart valve development is regulated by complex interactions between different cardiac cell types and is subject to blood flow-driven forces. Recent work has begun to elucidate the important roles of developmental pathways, valve cell heterogeneity and hemodynamics in determining the structure and function of developing valves. Furthermore, this work has revealed that many key genetic pathways involved in cardiac valve development are also implicated in diseased valves. Here, we review recent discoveries that have furthered our understanding of the molecular, cellular and mechanosensitive mechanisms of valve development, and highlight new insights into congenital and acquired valve disease.
Collapse
Affiliation(s)
- Anna O'Donnell
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Katherine E Yutzey
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
44
|
Analysis of factor V in zebrafish demonstrates minimal levels needed for early hemostasis. Blood Adv 2020; 3:1670-1680. [PMID: 31167819 DOI: 10.1182/bloodadvances.2018029066] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 04/17/2019] [Indexed: 12/28/2022] Open
Abstract
In humans, coagulation factor V (FV) deficiency is a rare, clinically heterogeneous bleeding disorder, suggesting that genetic modifiers may contribute to disease expressivity. Zebrafish possess many distinct advantages including high fecundity, optical clarity, external development, and homology with the mammalian hemostatic system, features that make it ideal for genetic studies. Our aim was to study the role of FV in zebrafish through targeted mutagenesis and apply the model to the study of human F5 variants. CRISPR-mediated genome editing of the zebrafish f5 locus was performed, generating mutants homozygous for a 49 base pair deletion in exon 4. Thrombus formation secondary to vascular endothelial injury was absent in f5 -/- mutant embryos and larvae. Despite this severe hemostatic defect, homozygous mutants survived before succumbing to severe hemorrhage in adulthood. Human F5 variants of uncertain significance from patients with FV deficiency were evaluated, and the causative mutations identified and stratified by their ability to restore thrombus formation in larvae. Analysis of these novel mutations demonstrates variable residual FV function, with minimal activity being required to restore hemostasis in response to laser-induced endothelial injury. This in vivo evaluation may be beneficial for patients whose factor activity levels lack correlation with bleeding symptomatology, although limitations exist. Furthermore, homozygous mutant embryos tolerate what is a severe and lethal defect in mammals, suggesting the possibility of species-specific factors enabling survival, and allowing further study not possible in the mouse. Identification of these factors or other genetic modifiers could lead to novel therapeutic modalities.
Collapse
|
45
|
Coronary vessel formation in development and disease: mechanisms and insights for therapy. Nat Rev Cardiol 2020; 17:790-806. [PMID: 32587347 DOI: 10.1038/s41569-020-0400-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2020] [Indexed: 12/20/2022]
Abstract
The formation of new blood vessels after myocardial infarction (MI) is essential for the survival of existing and regenerated cardiac tissue. However, the extent of endogenous revascularization after MI is insufficient, and MI can often result in ventricular remodelling, progression to heart failure and premature death. The neutral results of numerous clinical trials that have evaluated the efficacy of angiogenic therapy to revascularize the infarcted heart reflect our poor understanding of the processes required to form a functional coronary vasculature. In this Review, we describe the latest advances in our understanding of the processes involved in coronary vessel formation, with mechanistic insights taken from developmental studies. Coronary vessels originate from multiple cellular sources during development and form through a number of distinct and carefully orchestrated processes. The ectopic reactivation of developmental programmes has been proposed as a new paradigm for regenerative medicine, therefore, a complete understanding of these processes is crucial. Furthermore, knowledge of how these processes differ between the embryonic and adult heart, and how they might be more closely recapitulated after injury are critical for our understanding of regenerative biology, and might facilitate the identification of tractable molecular targets to therapeutically promote neovascularization and regeneration of the infarcted heart.
Collapse
|
46
|
Gancz D, Perlmoter G, Yaniv K. Formation and Growth of Cardiac Lymphatics during Embryonic Development, Heart Regeneration, and Disease. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037176. [PMID: 31818858 DOI: 10.1101/cshperspect.a037176] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The lymphatic system plays crucial roles in regulating fluid homeostasis, immune surveillance, and lipid transport. As is in most of the body's organs, the heart possesses an extensive lymphatic network. Moreover, a robust lymphangiogenic response has been shown to take place following myocardial infarction, highlighting cardiac lymphatics as potential targets for therapeutic intervention. Yet, the unique molecular properties and functions of the heart's lymphatic system have only recently begun to be addressed. In this review, we discuss the mechanisms underlying the formation and growth of cardiac lymphatics during embryonic development and describe their characteristics across species. We further summarize recent findings highlighting diverse cellular origins for cardiac lymphatic endothelial cells and how they integrate to form a single functional lymphatic network. Finally, we outline novel therapeutic avenues aimed at enhancing lymphatic vessel formation and integrity following cardiac injury, which hold great promise for promoting healing of the infarcted heart.
Collapse
Affiliation(s)
- Dana Gancz
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Gal Perlmoter
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Karina Yaniv
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
47
|
Gómez-González M, Latorre E, Arroyo M, Trepat X. Measuring mechanical stress in living tissues. NATURE REVIEWS. PHYSICS 2020; 2:300-317. [PMID: 39867749 PMCID: PMC7617344 DOI: 10.1038/s42254-020-0184-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/22/2020] [Indexed: 01/28/2025]
Abstract
Living tissues are active multifunctional materials capable of generating, sensing, withstanding and responding to mechanical stress. These capabilities enable tissues to adopt complex shapes during development, to sustain those shapes during homeostasis, and to restore them during healing and regeneration. Abnormal stress is associated with a broad range of pathologies, including developmental defects, inflammatory diseases, tumor growth and metastasis. Here we review techniques that measure mechanical stress in living tissues with cellular and subcellular resolution. We begin with 2D techniques to map stress in cultured cell monolayers, which provide the highest resolution and accessibility. These techniques include 2D traction microscopy, micro-pillar arrays, monolayer stress microscopy, and monolayer stretching between flexible cantilevers. We next focus on 3D traction microscopy and the micro-bulge test, which enable mapping forces in tissues cultured in 3D. Finally, we review techniques to measure stress in vivo, including servo-null methods for measuring luminal pressure, deformable inclusions, FRET sensors, laser ablation and computational methods for force inference. Whereas these techniques remain far from becoming everyday tools in biomedical laboratories, their rapid development is fostering key advances in the way we understand the role of mechanics in morphogenesis, homeostasis and disease.
Collapse
Affiliation(s)
- Manuel Gómez-González
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Ernest Latorre
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- LaCàN, Universitat Politècnica de Catalunya-BarcelonaTech, Barcelona, Spain
| | - Marino Arroyo
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- LaCàN, Universitat Politècnica de Catalunya-BarcelonaTech, Barcelona, Spain
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Barcelona08028, Spain
- Unitat de Biofísica i Bioenginyeria, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
48
|
Cardiac regeneration as an environmental adaptation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118623. [DOI: 10.1016/j.bbamcr.2019.118623] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/02/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022]
|
49
|
Advanced blood flow assessment in Zebrafish via experimental digital particle image velocimetry and computational fluid dynamics modeling. Micron 2020; 130:102801. [DOI: 10.1016/j.micron.2019.102801] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/18/2019] [Accepted: 12/13/2019] [Indexed: 01/23/2023]
|
50
|
Abstract
The hallmark of most cardiac diseases is the progressive loss of cardiomyocytes. In the perinatal period, cardiomyocytes still proliferate, and the heart shows the capacity to regenerate upon injury. In the adult heart, however, the actual rate of cardiomyocyte renewal is too low to efficiently counteract substantial cell loss caused by cardiac injury. In mammals, cardiac growth by cell number expansion changes to growth by cardiomyocyte enlargement soon after birth, coinciding with a period in which most cardiomyocytes increase their DNA content by multinucleation and nuclear polyploidization. Although cardiomyocyte hypertrophy is often associated with these processes, whether polyploidy is a prerequisite or a consequence of hypertrophic growth is unclear. Both the benefits of cardiomyocyte enlargement over proliferative growth of the heart and the physiological role of polyploidy in cardiomyocytes are enigmatic. Interestingly, hearts in animal species with substantial cardiac regenerative capacity dominantly comprise diploid cardiomyocytes, raising the hypothesis that cardiomyocyte polyploidy poses a barrier for cardiomyocyte proliferation and subsequent heart regeneration. On the contrary, there is also evidence for self-duplication of multinucleated myocytes, suggesting a more complex picture of polyploidy in heart regeneration. Polyploidy is not restricted to the heart but also occurs in other cell types in the body. In this review, we explore the biological relevance of polyploidy in different species and tissues to acquire insight into its specific role in cardiomyocytes. Furthermore, we speculate about the physiological role of polyploidy in cardiomyocytes and how this might relate to renewal and regeneration.
Collapse
Affiliation(s)
- Wouter Derks
- From the Center for Regenerative Therapies Dresden, Technische Universität Dresden, Germany (W.D., O.B.)
| | - Olaf Bergmann
- From the Center for Regenerative Therapies Dresden, Technische Universität Dresden, Germany (W.D., O.B.).,Karolinska Institutet, Cell and Molecular Biology, Stockholm, Sweden (O.B.)
| |
Collapse
|