51
|
Huang NC, Lee CM, Hsu SH. Effective naked plasmid DNA delivery into stem cells by microextrusion-based transient-transfection system for in situ cardiac repair. Cytotherapy 2020; 22:70-81. [PMID: 32007417 DOI: 10.1016/j.jcyt.2019.12.003] [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: 08/28/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 01/14/2023]
Abstract
BACKGROUND AIMS Combining the use of transfection reagents and physical methods can markedly improve the efficiency of gene delivery; however, such methods often cause cell damage. Additionally, naked plasmids without any vector or physical stimulation are difficult to deliver into stem cells. In this study, we demonstrate a simple and rapid method to simultaneously facilitate efficient in situ naked gene delivery and form a bioactive hydrogel scaffold. METHODS Transfecting naked GATA binding protein 4 (GATA4) plasmids into human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) by co-extruding naked plasmids and hUC-MSCs with a biomimetic and negatively charged water-based biodegradable thermo-responsive polyurethane (PU) hydrogel through a microextrusion-based transient-transfection system can upregulate the other cardiac marker genes. RESULTS The PU hydrogels with optimized physicochemical properties (such as hard-soft segment composition, size, hardness and thermal gelation) induced GATA4-transfected hUC-MSCs to express the cardiac marker proteins and then differentiated into cardiomyocyte-like cells in 15 days. We further demonstrated that GATA4-transfected hUC-MSCs in PU hydrogel were capable of in situ revival of heart function in zebrafish in 30 days. CONCLUSIONS Our results suggest that hUC-MSCs and naked plasmids encapsulated in PU hydrogels might represent a new strategy for in situ tissue therapy using the microextrusion-based transient-transfection system described here. This transfection system is simple, effective and safer than conventional technologies.
Collapse
Affiliation(s)
- Nien-Chi Huang
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, R.O.C
| | - Chii-Ming Lee
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, R.O.C
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, R.O.C.; Center of Tissue Engineering and 3D Printing, National Taiwan University, Taipei, Taiwan, R.O.C.; Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C..
| |
Collapse
|
52
|
De Spiegelaere W, Caboor L, Van Impe M, Boone MN, De Backer J, Segers P, Sips P. Corrosion casting of the cardiovascular structure in adult zebrafish for analysis by scanning electron microscopy and X-ray microtomography. Anat Histol Embryol 2020; 49:635-642. [PMID: 31995240 DOI: 10.1111/ahe.12535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/28/2019] [Accepted: 01/11/2020] [Indexed: 01/03/2023]
Abstract
Zebrafish have come to the forefront as a flexible, relevant animal model to study human disease, including cardiovascular disorders. Zebrafish are optically transparent during early developmental stages, enabling unparalleled imaging modalities to examine cardiovascular structure and function in vivo and ex vivo. At later stages, however, the options for systematic cardiovascular phenotyping are more limited. To visualise the complete vascular tree of adult zebrafish, we have optimised a vascular corrosion casting method. We present several improvements to the technique leading to increased reproducibility and accuracy. We designed a customised support system and used a combination of the commercially available Mercox II methyl methacrylate with the Batson's catalyst for optimal vascular corrosion casting of zebrafish. We also highlight different imaging approaches, with a focus on scanning electron microscopy (SEM) and X-ray microtomography (micro-CT) to obtain highly detailed, faithful three-dimensional reconstructed images of the zebrafish cardiovascular structure. This procedure can be of great value to a wide range of research lines related to cardiovascular biology in small specimens.
Collapse
Affiliation(s)
- Ward De Spiegelaere
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Lisa Caboor
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Matthias Van Impe
- Biofluid, Tissue and Solid Mechanics for Medical Applications (bioMMeda), Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Matthieu N Boone
- Center for X-ray Tomography, Department of Physics and Astronomy, Ghent University, Gent, Belgium
| | - Julie De Backer
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Patrick Segers
- Biofluid, Tissue and Solid Mechanics for Medical Applications (bioMMeda), Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Patrick Sips
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
53
|
Bensimon-Brito A, Ramkumar S, Boezio GLM, Guenther S, Kuenne C, Helker CSM, Sánchez-Iranzo H, Iloska D, Piesker J, Pullamsetti S, Mercader N, Beis D, Stainier DYR. TGF-β Signaling Promotes Tissue Formation during Cardiac Valve Regeneration in Adult Zebrafish. Dev Cell 2019; 52:9-20.e7. [PMID: 31786069 DOI: 10.1016/j.devcel.2019.10.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/17/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022]
Abstract
Cardiac valve disease can lead to severe cardiac dysfunction and is thus a frequent cause of morbidity and mortality. Its main treatment is valve replacement, which is currently greatly limited by the poor recellularization and tissue formation potential of the implanted valves. As we still lack suitable animal models to identify modulators of these processes, here we used adult zebrafish and found that, upon valve decellularization, they initiate a rapid regenerative program that leads to the formation of new functional valves. After injury, endothelial and kidney marrow-derived cells undergo cell cycle re-entry and differentiate into new extracellular matrix-secreting valve cells. The TGF-β signaling pathway promotes the regenerative process by enhancing progenitor cell proliferation as well as valve cell differentiation. These findings reveal a key role for TGF-β signaling in cardiac valve regeneration and establish the zebrafish as a model to identify and test factors promoting cardiac valve recellularization and growth.
Collapse
Affiliation(s)
- Anabela Bensimon-Brito
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany.
| | - Srinath Ramkumar
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Giulia L M Boezio
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Stefan Guenther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Carsten Kuenne
- Bioinformatics Core Unit, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Christian S M Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Héctor Sánchez-Iranzo
- Cell Biology and Biophysics Research Unit, EMBL Heidelberg, Heidelberg 69117, Germany
| | - Dijana Iloska
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Janett Piesker
- Scientific Service Group Microscopy, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Soni Pullamsetti
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Nadia Mercader
- Institute of Anatomy, University of Bern, Bern 3012, Switzerland; Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid 28049, Spain
| | - Dimitris Beis
- Developmental Biology, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany.
| |
Collapse
|
54
|
Pott A, Rottbauer W, Just S. Streamlining drug discovery assays for cardiovascular disease using zebrafish. Expert Opin Drug Discov 2019; 15:27-37. [PMID: 31570020 DOI: 10.1080/17460441.2020.1671351] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: In the last decade, our armamentarium of cardiovascular drug therapy has expanded significantly. Using innovative functional genomics strategies such as genome editing by CRISPR/Cas9 as well as high-throughput assays to identify bioactive small chemical compounds has significantly facilitated elaboration of the underlying pathomechanism in various cardiovascular diseases. However, despite scientific progress approvals for cardiovascular drugs has stagnated significantly compared to other fields of drug discovery and therapy during the past years.Areas covered: In this review, the authors discuss the aspects and pitfalls during the early phase of cardiovascular drug discovery and describe the advantages of zebrafish as an in vivo organism to model human cardiovascular diseases (CVD) as well as an in vivo platform for high-throughput chemical compound screening. They also highlight the emerging, promising techniques of automated read-out systems during high-throughput screening (HTS) for the evaluation of important cardiac functional parameters in zebrafish with the potential to streamline CVD drug discovery.Expert opinion: The successful identification of novel drugs to treat CVD is a major challenge in modern biomedical and clinical research. In this context, the definition of the etiologic fundamentals of human cardiovascular diseases is the prerequisite for an efficient and straightforward drug discovery.
Collapse
Affiliation(s)
- Alexander Pott
- Internal Medicine II, Ulm University Medical Center, Ulm, Germany.,Molecular Cardiology, Internal Medicine II, Ulm University Medical Center, Ulm, Germany
| | | | - Steffen Just
- Molecular Cardiology, Internal Medicine II, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
55
|
Abstract
Heart failure is a major cause of death worldwide owing to the inability of the adult human heart to regenerate after a heart attack. However, many vertebrate species are capable of complete cardiac regeneration following injury. In this Review, we discuss the various model organisms of cardiac regeneration, and outline what they have taught us thus far about the cellular and molecular responses essential for optimal cardiac repair. We compare across different species, highlighting evolutionarily conserved mechanisms of regeneration and demonstrating the importance of developmental gene expression programmes, plasticity of the heart and the pathophysiological environment for the regenerative response. Additionally, we discuss how the findings from these studies have led to improvements in cardiac repair in preclinical models such as adult mice and pigs, and discuss the potential to translate these findings into therapeutic approaches for human patients following myocardial infarction.
Collapse
Affiliation(s)
- Eleanor L Price
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Joaquim M Vieira
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Paul R Riley
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| |
Collapse
|
56
|
Vegfc/d-dependent regulation of the lymphatic vasculature during cardiac regeneration is influenced by injury context. NPJ Regen Med 2019; 4:18. [PMID: 31452940 PMCID: PMC6706389 DOI: 10.1038/s41536-019-0079-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 07/30/2019] [Indexed: 12/18/2022] Open
Abstract
The lymphatic vasculature mediates essential physiological functions including fluid homeostasis, lipid and hormone transport, and immune cell trafficking. Recent studies have suggested that promoting lymphangiogenesis enhances cardiac repair following injury, but it is unknown whether lymphangiogenesis is required for cardiac regeneration. Here, we describe the anatomical distribution, regulation, and function of the cardiac lymphatic network in a highly regenerative zebrafish model system using transgenic reporter lines and loss-of-function approaches. We show that zebrafish lacking functional vegfc and vegfd signaling are devoid of a cardiac lymphatic network and display cardiac hypertrophy in the absence of injury, suggesting a role for these vessels in cardiac tissue homeostasis. Using two different cardiac injury models, we report a robust lymphangiogenic response following cryoinjury, but not following apical resection injury. Although the majority of mutants lacking functional vegfc and vegfd signaling were able to mount a full regenerative response even in the complete absence of a cardiac lymphatic vasculature, cardiac regeneration was severely impaired in a subset of mutants, which was associated with heightened pro-inflammatory cytokine signaling. These findings reveal a context-dependent requirement for the lymphatic vasculature during cardiac growth and regeneration.
Collapse
|
57
|
Narumanchi S, Kalervo K, Perttunen S, Wang H, Immonen K, Kosonen R, Laine M, Ruskoaho H, Tikkanen I, Lakkisto P, Paavola J. Inhibition of let-7c Regulates Cardiac Regeneration after Cryoinjury in Adult Zebrafish. J Cardiovasc Dev Dis 2019; 6:jcdd6020016. [PMID: 30987331 PMCID: PMC6617397 DOI: 10.3390/jcdd6020016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/30/2019] [Accepted: 04/01/2019] [Indexed: 12/12/2022] Open
Abstract
The let-7c family of micro-RNAs (miRNAs) is expressed during embryonic development and plays an important role in cell differentiation. We have investigated the role of let-7c in heart regeneration after injury in adult zebrafish. let-7c antagomir or scramble injections were given at one day after cryoinjury (1 dpi). Tissue samples were collected at 7 dpi, 14 dpi and 28 dpi and cardiac function was assessed before cryoinjury, 1 dpi, 7 dpi, 14 dpi and 28 dpi. Inhibition of let-7c increased the rate of fibrinolysis, increased the number of proliferating cell nuclear antigen (PCNA) positive cardiomyocytes at 7 dpi and increased the expression of the epicardial marker raldh2 at 7 dpi. Additionally, cardiac function measured with echocardiography recovered slightly more rapidly after inhibition of let-7c. These results reveal a beneficial role of let-7c inhibition in adult zebrafish heart regeneration.
Collapse
Affiliation(s)
- Suneeta Narumanchi
- Unit of Cardiovascular Research, Minerva Institute for Medical Research, Biomedicum Helsinki, 00290 Helsinki, Finland.
| | - Karri Kalervo
- Unit of Cardiovascular Research, Minerva Institute for Medical Research, Biomedicum Helsinki, 00290 Helsinki, Finland.
- Department of Surgery, South Karelia Central Hospital, 53130 Lappeenranta, Finland.
| | - Sanni Perttunen
- Unit of Cardiovascular Research, Minerva Institute for Medical Research, Biomedicum Helsinki, 00290 Helsinki, Finland.
| | - Hong Wang
- Unit of Cardiovascular Research, Minerva Institute for Medical Research, Biomedicum Helsinki, 00290 Helsinki, Finland.
| | - Katariina Immonen
- Unit of Cardiovascular Research, Minerva Institute for Medical Research, Biomedicum Helsinki, 00290 Helsinki, Finland.
| | - Riikka Kosonen
- Unit of Cardiovascular Research, Minerva Institute for Medical Research, Biomedicum Helsinki, 00290 Helsinki, Finland.
| | - Mika Laine
- Unit of Cardiovascular Research, Minerva Institute for Medical Research, Biomedicum Helsinki, 00290 Helsinki, Finland.
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland.
| | - Heikki Ruskoaho
- Drug Research Programme, Division of Pharmacology and Pharmacotherapy, University of Helsinki, 00014 Helsinki, Finland.
| | - Ilkka Tikkanen
- Unit of Cardiovascular Research, Minerva Institute for Medical Research, Biomedicum Helsinki, 00290 Helsinki, Finland.
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland.
| | - Päivi Lakkisto
- Unit of Cardiovascular Research, Minerva Institute for Medical Research, Biomedicum Helsinki, 00290 Helsinki, Finland.
- Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital, 00014 Helsinki, Finland.
| | - Jere Paavola
- Unit of Cardiovascular Research, Minerva Institute for Medical Research, Biomedicum Helsinki, 00290 Helsinki, Finland.
- Clinical Neurosciences, Neurology, University of Helsinki and Jorvi Hospital of Helsinki University Hospital, 02740 Espoo, Finland.
| |
Collapse
|
58
|
Li YF, Canário AVM, Power DM, Campinho MA. Ioxynil and diethylstilbestrol disrupt vascular and heart development in zebrafish. ENVIRONMENT INTERNATIONAL 2019; 124:511-520. [PMID: 30685453 DOI: 10.1016/j.envint.2019.01.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 12/17/2018] [Accepted: 01/04/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Endocrine disruption is one of the consequences of industrialization and chemicals released into the environment have a profound impact on organisms. Waterborne micromolar concentrations of ioxynil (IOX) and diethylstilbestrol (DES) in fish affect the development of the heart, vasculature and thyroid gland. OBJECTIVES The present study aimed to determine how IOX and DES disrupt the crosstalk between the developing thyroid gland and cardio-vascular system in zebrafish. METHODS Twelve hours post fertilization (hpf) wild type, Tg(fli1:GFP) or Tg(cmalc2:GFPCaaX) zebrafish embryos were exposed to 0.1 μM IOX or DES for 36 h (up until 48 hpf) or 60 h (up until 72 hpf). Embryos were used for vascular endothelial cell sorting, whole-mount immunohistochemistry, tissue selective transcriptomics, selected gene expression analysis by quantitative real-time polymerase chain reaction analysis and determination of heart rate by live imaging. RESULTS Exposure of zebrafish embryos to IOX and DES (0.1 μM) increased heart beat frequency and reduced ventricle volume and aorta diameter. The transcriptome of endothelial cells from blood vessels of hypertrophic, dilated and arrhythmogenic right ventricular cardiomyopathy was significantly changed and compound-specific toxic effects were found in IOX and DES exposed embryos. Both DES and IOX directly affected vascular and heart development and this indirectly impaired thyroid gland development in zebrafish. Even though the toxicity end-point of the two chemicals was similar, their action seemed to be via different gene regulatory pathways and physiological mechanisms. CONCLUSION IOX and DES directly disrupt cardiovascular development and there is an associated disruption of thyroid tissue that most likely has long term consequences for this endocrine axis.
Collapse
Affiliation(s)
- Yi-Feng Li
- International Research Centre for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China; Centre of Marine Sciences, University of Algarve, Faro, Portugal
| | - Adelino V M Canário
- International Research Centre for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China; Centre of Marine Sciences, University of Algarve, Faro, Portugal
| | - Deborah M Power
- International Research Centre for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China; Centre of Marine Sciences, University of Algarve, Faro, Portugal
| | - Marco A Campinho
- Centre of Marine Sciences, University of Algarve, Faro, Portugal.
| |
Collapse
|
59
|
Schulz A, Brendler J, Blaschuk O, Landgraf K, Krueger M, Ricken AM. Non-pathological Chondrogenic Features of Valve Interstitial Cells in Normal Adult Zebrafish. J Histochem Cytochem 2019; 67:361-373. [PMID: 30620237 DOI: 10.1369/0022155418824083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
In the heart, unidirectional blood flow depends on proper heart valve function. As, in mammals, regulatory mechanisms of early heart valve and bone development are shown to contribute to adult heart valve pathologies, we used the animal model zebrafish (ZF, Danio rerio) to investigate the microarchitecture and differentiation of cardiac valve interstitial cells in the transition from juvenile (35 days) to end of adult breeding (2.5 years) stages. Of note, light microscopy and immunohistochemistry revealed major differences in ZF heart valve microarchitecture when compared with adult mice. We demonstrate evidence for rather chondrogenic features of valvular interstitial cells by histological staining and immunodetection of SOX-9, aggrecan, and type 2a1 collagen. Collagen depositions are enriched in a thin layer at the atrial aspect of atrioventricular valves and the ventricular aspect of bulboventricular valves, respectively. At the ultrastructural level, the collagen fibrils are lacking obvious periodicity and orientation throughout the entire valve.
Collapse
Affiliation(s)
- Alina Schulz
- Institute of Anatomy, Faculty of Medicine.,University of Leipzig, Leipzig, Germany
| | - Jana Brendler
- Institute of Anatomy, Faculty of Medicine.,University of Leipzig, Leipzig, Germany
| | - Orest Blaschuk
- Division of Urology, Department of Surgery, McGill University, Montreal, Québec, Canada.,University of Leipzig, Leipzig, Germany
| | - Kathrin Landgraf
- Center for Pediatric Research Leipzig, University Hospital for Children & Adolescents and Integrated Research and Treatment Centre Adiposity Diseases.,University of Leipzig, Leipzig, Germany
| | - Martin Krueger
- Institute of Anatomy, Faculty of Medicine.,University of Leipzig, Leipzig, Germany
| | - Albert M Ricken
- Institute of Anatomy, Faculty of Medicine.,University of Leipzig, Leipzig, Germany
| |
Collapse
|
60
|
Monteiro DA, Kalinin AL, Selistre-de-Araújo HS, Nogueira LAN, Beletti ME, Fernandes MN, Rantin FT. Cardioprotective effects of alternagin-C (ALT-C), a disintegrin-like protein from Rhinocerophis alternatus snake venom, on hypoxia-reoxygenation-induced injury in fish. Comp Biochem Physiol C Toxicol Pharmacol 2019; 215:67-75. [PMID: 30352307 DOI: 10.1016/j.cbpc.2018.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 10/10/2018] [Indexed: 10/28/2022]
Abstract
Alternagin-C (ALT-C) is a disintegrin-like peptide purified from Rhinocerophis alternatus snake venom with the property of inducing vascular endothelial growth factor (VEGF) expression, endothelial cell proliferation and migration, and angiogenesis. Therefore, this protein could be interesting as a new approach for ischemic heart diseases, an imbalance between myocardial oxygen supply and demand, leading to cardiac dysfunction. We investigated the effects of a single dose of alternagin-C (0.5 mg kg-1, via intra-arterial), after 7 days, on hypoxia/reoxygenation challenge in isolated ventricle strips and on morphological changes and density of blood vessels of the heart, using fish as an alternative experimental model. ALT-C treatment provided protection of cardiomyocytes against hypoxia/reoxygenation-induced negative inotropism. ALT-C also stimulated angiogenesis and improved excitation-contraction coupling during hypoxic conditions. Our results provide a new insight into a functional role of ALT-C against hypoxia/reoxygenation-induced cardiomyocyte injury pointing out to a potential therapeutic strategy for ischemia-related diseases.
Collapse
Affiliation(s)
- D A Monteiro
- Department of Physiological Sciences, Federal University of São Carlos, São Carlos, São Paulo, Brazil.
| | - A L Kalinin
- Department of Physiological Sciences, Federal University of São Carlos, São Carlos, São Paulo, Brazil
| | - H S Selistre-de-Araújo
- Department of Physiological Sciences, Federal University of São Carlos, São Carlos, São Paulo, Brazil
| | - L A N Nogueira
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - M E Beletti
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - M N Fernandes
- Department of Physiological Sciences, Federal University of São Carlos, São Carlos, São Paulo, Brazil
| | - F T Rantin
- Department of Physiological Sciences, Federal University of São Carlos, São Carlos, São Paulo, Brazil
| |
Collapse
|
61
|
Cui M, Wang Z, Bassel-Duby R, Olson EN. Genetic and epigenetic regulation of cardiomyocytes in development, regeneration and disease. Development 2018; 145:145/24/dev171983. [PMID: 30573475 DOI: 10.1242/dev.171983] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Embryonic and postnatal life depend on the uninterrupted function of cardiac muscle cells. These cells, termed cardiomyocytes, display many fascinating behaviors, including complex morphogenic movements, interactions with other cell types of the heart, persistent contractility and quiescence after birth. Each of these behaviors depends on complex interactions between both cardiac-restricted and widely expressed transcription factors, as well as on epigenetic modifications. Here, we review recent advances in our understanding of the genetic and epigenetic control of cardiomyocyte differentiation and proliferation during heart development, regeneration and disease. We focus on those regulators that are required for both heart development and disease, and highlight the regenerative principles that might be manipulated to restore function to the injured adult heart.
Collapse
Affiliation(s)
- Miao Cui
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Zhaoning Wang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Eric N Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| |
Collapse
|
62
|
Stoyek MR, Rog-Zielinska EA, Quinn TA. Age-associated changes in electrical function of the zebrafish heart. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 138:91-104. [DOI: 10.1016/j.pbiomolbio.2018.07.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/24/2018] [Accepted: 07/25/2018] [Indexed: 12/17/2022]
|
63
|
Ultracytochemical visualization of calcium distribution in heart cells and erythrocytes of zebrafish Danio rerio. Micron 2018; 111:19-27. [DOI: 10.1016/j.micron.2018.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 05/23/2018] [Accepted: 05/23/2018] [Indexed: 01/06/2023]
|
64
|
Restoration of cardiac function after anaemia-induced heart failure in zebrafish. J Mol Cell Cardiol 2018; 121:223-232. [PMID: 30009777 DOI: 10.1016/j.yjmcc.2018.07.128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 06/28/2018] [Accepted: 07/11/2018] [Indexed: 01/22/2023]
Abstract
AIMS New therapeutic approaches are needed to fight against the growing epidemic of heart failure. Unlike mammals, zebrafish possess the incredible ability to regenerate cardiac tissue after acute trauma such as apical resection. Yet, the ability of zebrafish to recover after a chronic stress leading to heart failure has not been reported. The aim of this study was to test whether zebrafish can recover a normal cardiac function after anaemia-induced heart failure. METHODS AND RESULTS Eight- to ten-month-old zebrafish were treated with phenylhydrazine hydrochloride, an anaemia inducer, to generate heart failure. Treatment was stopped after 5 weeks and fish were followed-up for 3 weeks. Assessment of ventricular function by ultrasound at the end of the treatment revealed an increase in ventricle diameter (+47%) and a decrease in heart rate (-36%) and fractional shortening (-30%). A decrease in swim capacity was also observed (-31%). Tissue staining showed a thickening of the ventricular wall (5-fold), cell apoptosis and proliferation but no fibrosis. Expression of foetal genes, angiogenic factor and inflammation markers was increased, and β-adrenergic receptor-1 was decreased. Three weeks after phenylhydrazine hydrochloride withdrawal, all parameters returned to baseline and the fish recovered a normal cardiac function, tissue morphology and gene expression. CONCLUSIONS Zebrafish are able to completely recover from anaemia-induced heart failure. This model represents a unique opportunity to investigate the mechanisms of cardiac repair and may lead to the discovery of novel therapeutic targets of heart failure.
Collapse
|
65
|
Choi W, Kim HM, Park S, Yeom E, Doh J, Lee SJ. Variation in wall shear stress in channel networks of zebrafish models. J R Soc Interface 2017; 14:rsif.2016.0900. [PMID: 28148768 DOI: 10.1098/rsif.2016.0900] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/09/2017] [Indexed: 01/13/2023] Open
Abstract
Physiological functions of vascular endothelial cells (ECs) vary depending on wall shear stress (WSS) magnitude, and the functional change affects the pathologies of various cardiovascular systems. Several in vitro and in vivo models have been used to investigate the functions of ECs under different WSS conditions. However, these models have technical limitations in precisely mimicking the physiological environments of ECs and monitoring temporal variations of ECs in detail. Although zebrafish (Danio rerio) has several strategies to overcome these technical limitations, zebrafish cannot be used as a perfect animal model because applying various WSS conditions on blood vessels of zebrafish is difficult. This study proposes a new zebrafish model in which various WSS can be applied to the caudal vein. The WSS magnitude is controlled by blocking some parts of blood-vessel networks. The accuracy and reproducibility of the proposed method are validated using an equivalent circuit model of blood vessels in zebrafish. The proposed method is applied to lipopolysaccharide (LPS)-stimulated zebrafish as a typical application. The proposed zebrafish model can be used as an in vivo animal model to investigate the relationship between WSS and EC physiology or WSS-induced cardiovascular diseases.
Collapse
Affiliation(s)
- Woorak Choi
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), San 31, Hyoja-dong, Pohang 790-784, South Korea
| | - Hye Mi Kim
- Division of Integrative Biosciences and Biotechnology (IBB), Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Sungho Park
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), San 31, Hyoja-dong, Pohang 790-784, South Korea
| | - Eunseop Yeom
- School of Mechanical Engineering, Pusan National University, Busan, South Korea
| | - Junsang Doh
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), San 31, Hyoja-dong, Pohang 790-784, South Korea
| | - Sang Joon Lee
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), San 31, Hyoja-dong, Pohang 790-784, South Korea
| |
Collapse
|
66
|
Sun Y, Wang Q, Fang Y, Wu C, Lu G, Chen Z. Activation of the Nkx2.5-Calr-p53 signaling pathway by hyperglycemia induces cardiac remodeling and dysfunction in adult zebrafish. Dis Model Mech 2017; 10:1217-1227. [PMID: 28801532 PMCID: PMC5665450 DOI: 10.1242/dmm.026781] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 07/31/2017] [Indexed: 01/19/2023] Open
Abstract
Hyperglycemia is an independent risk factor for diabetic cardiomyopathy in humans; however, the underlying mechanisms have not been thoroughly elucidated. Zebrafish (Danio rerio) was used in this study as a novel vertebrate model to explore the signaling pathways of human adult cardiomyopathy. Hyperglycemia was induced by alternately immersing adult zebrafish in a glucose solution or water. The hyperglycemic fish gradually exhibited some hallmarks of cardiomyopathy such as myocardial hypertrophy and apoptosis, myofibril loss, fetal gene reactivation, and severe arrhythmia. Echocardiography of the glucose-treated fish demonstrated diastolic dysfunction at an early stage and systolic dysfunction at a later stage, consistent with what is observed in diabetic patients. Enlarged hearts with decreased myocardial density, accompanied by decompensated cardiac function, indicated that apoptosis was critical in the pathological process. Significant upregulation of the expression of Nkx2.5 and its downstream targets calreticulin (Calr) and p53 was noted in the glucose-treated fish. High-glucose stimulation in vitro evoked marked apoptosis of primary cardiomyocytes, which was rescued by the p53 inhibitor pifithrin-μ. In vitro experiments were performed using compound treatment and genetically via cell infection. Genetically, knockout of Nkx2.5 induced decreased expression of Nkx2.5, Calr and p53 Upregulation of Calr resulted in increased p53 expression, whereas the level of Nkx2.5 remained unchanged. An adult zebrafish model of hyperglycemia-induced cardiomyopathy was successfully established. Hyperglycemia-induced myocardial apoptosis was mediated, at least in part, by activation of the Nkx2.5-Calr-p53 pathway in vivo, resulting in cardiac dysfunction and hyperglycemia-induced cardiomyopathy.
Collapse
Affiliation(s)
- Yanyi Sun
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197, Ruijin Er Road, Shanghai, 200025, China
| | - Qiuyun Wang
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197, Ruijin Er Road, Shanghai, 200025, China
| | - Yuehua Fang
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197, Ruijin Er Road, Shanghai, 200025, China
| | - Chunfang Wu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197, Ruijin Er Road, Shanghai, 200025, China
| | - Guoping Lu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197, Ruijin Er Road, Shanghai, 200025, China
| | - Zhenyue Chen
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197, Ruijin Er Road, Shanghai, 200025, China
| |
Collapse
|
67
|
MacDonald EA, Stoyek MR, Rose RA, Quinn TA. Intrinsic regulation of sinoatrial node function and the zebrafish as a model of stretch effects on pacemaking. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017; 130:198-211. [PMID: 28743586 DOI: 10.1016/j.pbiomolbio.2017.07.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 07/17/2017] [Accepted: 07/21/2017] [Indexed: 12/18/2022]
Abstract
Excitation of the heart occurs in a specialised region known as the sinoatrial node (SAN). Tight regulation of SAN function is essential for the maintenance of normal heart rhythm and the response to (patho-)physiological changes. The SAN is regulated by extrinsic (central nervous system) and intrinsic (neurons, peptides, mechanics) factors. The positive chronotropic response to stretch in particular is essential for beat-by-beat adaptation to changes in hemodynamic load. Yet, the mechanism of this stretch response is unknown, due in part to the lack of an appropriate experimental model for targeted investigations. We have been investigating the zebrafish as a model for the study of intrinsic regulation of SAN function. In this paper, we first briefly review current knowledge of the principal components of extrinsic and intrinsic SAN regulation, derived primarily from experiments in mammals, followed by a description of the zebrafish as a novel experimental model for studies of intrinsic SAN regulation. This mini-review is followed by an original investigation of the response of the zebrafish isolated SAN to controlled stretch. Stretch causes an immediate and continuous increase in beating rate in the zebrafish isolated SAN. This increase reaches a maximum part way through a period of sustained stretch, with the total change dependent on the magnitude and direction of stretch. This is comparable to what occurs in isolated SAN from most mammals (including human), suggesting that the zebrafish is a novel experimental model for the study of mechanisms involved in the intrinsic regulation of SAN function by mechanical effects.
Collapse
Affiliation(s)
- Eilidh A MacDonald
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Matthew R Stoyek
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Robert A Rose
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada; School of Biomedical Engineering, Dalhousie University, Halifax, Canada.
| |
Collapse
|
68
|
Cahill TJ, Choudhury RP, Riley PR. Heart regeneration and repair after myocardial infarction: translational opportunities for novel therapeutics. Nat Rev Drug Discov 2017; 16:699-717. [DOI: 10.1038/nrd.2017.106] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
69
|
The zebrafish ventricle: A hub of cardiac endothelial cells for in vitro cell behavior studies. Sci Rep 2017; 7:2687. [PMID: 28578380 PMCID: PMC5457396 DOI: 10.1038/s41598-017-02461-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 04/12/2017] [Indexed: 11/09/2022] Open
Abstract
Despite our increasing understanding of zebrafish heart development and regeneration, there is limited information about the distribution of endothelial cells (ECs) in the adult zebrafish heart. Here, we investigate and compare the distribution of cardiac ECs (cECs) in adult mouse and zebrafish ventricles. Surprisingly, we find that (i) active coronary vessel growth is present in adult zebrafish, (ii) ~37 and ~39% of cells in the zebrafish heart are ECs and cardiomyocytes, respectively, a composition similar to that seen in mouse. However, we find that in zebrafish, ~36% of the ventricular tissue is covered with ECs, i.e., a substantially larger proportion than in mouse. Capitalising on the high abundance of cECs in zebrafish, we established a protocol to isolate them with high purity using fluorescent transgenic lines. Our approach eliminates side-effects due to antibody utilisation. Moreover, the isolated cECs maintained a high proliferation index even after three passages and were amenable to pharmacological treatments to study cEC migration in vitro. Such primary cultures will be a useful tool for supplementary in vitro studies on the accumulating zebrafish mutant lines as well as the screening of small molecule libraries on cardiac specific endothelial cells.
Collapse
|
70
|
Genome editing of factor X in zebrafish reveals unexpected tolerance of severe defects in the common pathway. Blood 2017; 130:666-676. [PMID: 28576875 DOI: 10.1182/blood-2017-02-765206] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/23/2017] [Indexed: 12/24/2022] Open
Abstract
Deficiency of factor X (F10) in humans is a rare bleeding disorder with a heterogeneous phenotype and limited therapeutic options. Targeted disruption of F10 and other common pathway factors in mice results in embryonic/neonatal lethality with rapid resorption of homozygous mutants, hampering additional studies. Several of these mutants also display yolk sac vascular defects, suggesting a role for thrombin signaling in vessel development. The zebrafish is a vertebrate model that demonstrates conservation of the mammalian hemostatic and vascular systems. We have leveraged these advantages for in-depth study of the role of the coagulation cascade in the developmental regulation of hemostasis and vasculogenesis. In this article, we show that ablation of zebrafish f10 by using genome editing with transcription activator-like effector nucleases results in a major embryonic hemostatic defect. However, widespread hemorrhage and subsequent lethality does not occur until later stages, with absence of any detectable defect in vascular development. We also use f10-/- zebrafish to confirm 5 novel human F10 variants as causative mutations in affected patients, providing a rapid and reliable in vivo model for testing the severity of F10 variants. These findings as well as the prolonged survival of f10-/- mutants will enable us to expand our understanding of the molecular mechanisms of hemostasis, including a platform for screening variants of uncertain significance in patients with F10 deficiency and other coagulation disorders. Further study as to how fish tolerate what is an early lethal mutation in mammals could facilitate improvement of diagnostics and therapeutics for affected patients with bleeding disorders.
Collapse
|
71
|
Pfefferli C, Jaźwińska A. The careg element reveals a common regulation of regeneration in the zebrafish myocardium and fin. Nat Commun 2017; 8:15151. [PMID: 28466843 PMCID: PMC5418624 DOI: 10.1038/ncomms15151] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 03/03/2017] [Indexed: 12/21/2022] Open
Abstract
The existence of common mechanisms regulating organ regeneration is an intriguing concept. Here we report on a regulatory element that is transiently activated during heart and fin regeneration in zebrafish. This element contains a ctgfa upstream sequence, called careg, which is induced by TGFβ/Activin-β signalling in the peri-injury zone of the myocardium and the fin mesenchyme. In addition, this reporter demarcates a primordial cardiac layer and intraray osteoblasts. Using genetic fate mapping, we show the regenerative competence of careg-expressing cells. The analysis of the heart reveals that the primordial cardiac layer is incompletely restored after cryoinjury, whereas trabecular and cortical cardiomyocytes contribute to myocardial regrowth. In regenerating fins, the activated mesenchyme of the stump gives rise to the blastema. Our findings provide evidence of a common regenerative programme in cardiomyocytes and mesenchyme that opens the possibility to further explore conserved mechanisms of the cellular plasticity in diverse vertebrate organs.
Collapse
Affiliation(s)
- Catherine Pfefferli
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| |
Collapse
|
72
|
Yalcin HC, Amindari A, Butcher JT, Althani A, Yacoub M. Heart function and hemodynamic analysis for zebrafish embryos. Dev Dyn 2017; 246:868-880. [PMID: 28249360 DOI: 10.1002/dvdy.24497] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/24/2017] [Accepted: 02/24/2017] [Indexed: 12/28/2022] Open
Abstract
The Zebrafish has emerged to become a powerful vertebrate animal model for cardiovascular research in recent years. Its advantages include easy genetic manipulation, transparency, small size, low cost, and the ability to survive without active circulation at early stages of development. Sequencing the whole genome and identifying ortholog genes with human genome made it possible to induce clinically relevant cardiovascular defects via genetic approaches. Heart function and disturbed hemodynamics need to be assessed in a reliable manner for these disease models in order to reveal the mechanobiology of induced defects. This effort requires precise determination of blood flow patterns as well as hemodynamic stress (i.e., wall shear stress and pressure) levels within the developing heart. While traditional approach involves time-lapse brightfield microscopy to track cell and tissue movements, in more recent studies fast light-sheet fluorescent microscopes are utilized for that purpose. Integration of more complicated techniques like particle image velocimetry and computational fluid dynamics modeling for hemodynamic analysis holds a great promise to the advancement of the Zebrafish studies. Here, we discuss the latest developments in heart function and hemodynamic analysis for Zebrafish embryos and conclude with our future perspective on dynamic analysis of the Zebrafish cardiovascular system. Developmental Dynamics 246:868-880, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Armin Amindari
- Faculty of Mechanical Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Jonathan T Butcher
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Asma Althani
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Magdi Yacoub
- Imperial College, NHLI, Heart Science Centre, Harefield, Middlesex, UB9 6JH, United Kingdom
| |
Collapse
|
73
|
Jiang XY, Sarsons CD, Gomez-Garcia MJ, Cramb DT, Rinker KD, Childs SJ. Quantum dot interactions and flow effects in angiogenic zebrafish ( Danio rerio ) vessels and human endothelial cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:999-1010. [DOI: 10.1016/j.nano.2016.12.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/21/2016] [Accepted: 12/05/2016] [Indexed: 01/21/2023]
|
74
|
Karra R, Poss KD. Redirecting cardiac growth mechanisms for therapeutic regeneration. J Clin Invest 2017; 127:427-436. [PMID: 28145902 DOI: 10.1172/jci89786] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Heart failure is a major source of morbidity and mortality. Replacing lost myocardium with new tissue is a major goal of regenerative medicine. Unlike adult mammals, zebrafish and neonatal mice are capable of heart regeneration following cardiac injury. In both contexts, the regenerative program echoes molecular and cellular events that occur during cardiac development and morphogenesis, notably muscle creation through division of cardiomyocytes. Based on studies over the past decade, it is now accepted that the adult mammalian heart undergoes a low grade of cardiomyocyte turnover. Recent data suggest that this cardiomyocyte turnover can be augmented in the adult mammalian heart by redeployment of developmental factors. These findings and others suggest that stimulating endogenous regenerative responses can emerge as a therapeutic strategy for human cardiovascular disease.
Collapse
|
75
|
Abstract
The pronephros is the first kidney type to form in vertebrate embryos. The first step of pronephrogenesis in the zebrafish is the formation of the intermediate mesoderm during gastrulation, which occurs in response to secreted morphogens such as BMPs and Nodals. Patterning of the intermediate mesoderm into proximal and distal cell fates is induced by retinoic acid signaling with downstream transcription factors including wt1a, pax2a, pax8, hnf1b, sim1a, mecom, and irx3b. In the anterior intermediate mesoderm, progenitors of the glomerular blood filter migrate and fuse at the midline and recruit a blood supply. More posteriorly localized tubule progenitors undergo epithelialization and fuse with the cloaca. The Notch signaling pathway regulates the formation of multi-ciliated cells in the tubules and these cells help propel the filtrate to the cloaca. The lumenal sheer stress caused by flow down the tubule activates anterior collective migration of the proximal tubules and induces stretching and proliferation of the more distal segments. Ultimately these processes create a simple two-nephron kidney that is capable of reabsorbing and secreting solutes and expelling excess water-processes that are critical to the homeostasis of the body fluids. The zebrafish pronephric kidney provides a simple, yet powerful, model system to better understand the conserved molecular and cellular progresses that drive nephron formation, structure, and function.
Collapse
Affiliation(s)
- Richard W Naylor
- Department of Molecular Medicine and Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Sarah S Qubisi
- Department of Molecular Medicine and Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Alan J Davidson
- Department of Molecular Medicine and Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
| |
Collapse
|
76
|
Garg R, Ko GM, Smith CJ. QSAR molecular parameters calculated for US EPA ToxCast Phase 1 and 2 chemical compounds tested against embryonic zebrafish. TOXICOLOGY RESEARCH AND APPLICATION 2017. [DOI: 10.1177/2397847317707371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Freshwater and marine environments are exposed to small concentrations of many different chemicals produced by industrial, agricultural, pharmaceutical, cosmetic, food, and household applications. Due to concerns regarding potential adverse events from these exposures, regulatory agencies around the world have established aquatic toxicology testing protocols that measure untoward responses in a wide variety of freshwater and marine organisms. Following a literature review of databases on the toxicity of chemicals to fish, the embryonic zebrafish ( Danio rerio) database compiled by the Tanguay Laboratory at Oregon State University was determined to be well suited for quantitative structure–activity relationship (QSAR) analysis. This database possesses a number of favorable characteristics including large size (1060 unique US Environmental Protection Agency ToxCast phase 1 and 2 chemical compounds), relatively recent data collected using state-of-the-art methods, 18 simultaneously measured toxicological end points, transparent embryos that develop externally thereby facilitating toxicological evaluation, and the vast majority of the genetic code is expressed and active during early life stages. The molecular parameters calculated for each of the chemicals in the database include the logarithm of the octanol–water partition coefficient, molar volume, and molar refractivity. For each chemical, the availability of these molecular parameter values can facilitate future QSAR studies using any of the 18 different toxicological end points measured as the biological activity of interest.
Collapse
Affiliation(s)
- Rajni Garg
- Computational Science Research Center, San Diego State University, San Diego, CA, USA
| | - Gene M Ko
- Electromagnetic Systems Group, General Atomics, San Diego, CA, USA
| | - Carr J Smith
- Department of Nurse Anesthesia, Florida State University, Panama City, FL, USA
| |
Collapse
|
77
|
Burggren WW, Dubansky B, Bautista NM. Cardiovascular Development in Embryonic and Larval Fishes. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/bs.fp.2017.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
78
|
|
79
|
Wang LW, Huttner IG, Santiago CF, Kesteven SH, Yu ZY, Feneley MP, Fatkin D. Standardized echocardiographic assessment of cardiac function in normal adult zebrafish and heart disease models. Dis Model Mech 2016; 10:63-76. [PMID: 28067629 PMCID: PMC5278526 DOI: 10.1242/dmm.026989] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 11/11/2016] [Indexed: 11/20/2022] Open
Abstract
The zebrafish (Danio rerio) is an increasingly popular model organism in cardiovascular research. Major insights into cardiac developmental processes have been gained by studies of embryonic zebrafish. However, the utility of zebrafish for modeling adult-onset heart disease has been limited by a lack of robust methods for in vivo evaluation of cardiac function. We established a physiological protocol for underwater zebrafish echocardiography using high frequency ultrasound, and evaluated its reliability in detecting altered cardiac function in two disease models. Serial assessment of cardiac function was performed in wild-type zebrafish aged 3 to 12 months and the effects of anesthetic agents, age, sex and background strain were evaluated. There was a varying extent of bradycardia and ventricular contractile impairment with different anesthetic drugs and doses, with tricaine 0.75 mmol l−1 having a relatively more favorable profile. When compared with males, female fish were larger and had more measurement variability. Although age-related increments in ventricular chamber size were greater in females than males, there were no sex differences when data were normalized to body size. Systolic ventricular function was similar in both sexes at all time points, but differences in diastolic function were evident from 6 months onwards. Wild-type fish of both sexes showed a reliance on atrial contraction for ventricular diastolic filling. Echocardiographic evaluation of adult zebrafish with diphtheria toxin-induced myocarditis or anemia-induced volume overload accurately identified ventricular dilation and altered contraction, with suites of B-mode, ventricular strain, pulsed-wave Doppler and tissue Doppler indices showing concordant changes indicative of myocardial hypocontractility or hypercontractility, respectively. Repeatability, intra-observer and inter-observer correlations for echocardiographic measurements were high. We demonstrate that high frequency echocardiography allows reliable in vivo cardiac assessment in adult zebrafish and make recommendations for optimizing data acquisition and analysis. This enabling technology reveals new insights into zebrafish cardiac physiology and provides an imaging platform for zebrafish-based translational research. Summary: Standardization of zebrafish echocardiography provides insights into cardiac physiology in normal and diseased states, with application for functional studies in zebrafish models of heart disease.
Collapse
Affiliation(s)
- Louis W Wang
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia.,Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2052, Australia.,Department of Cardiology, St Vincent's Hospital, Darlinghurst, New South Wales 2010, Australia
| | - Inken G Huttner
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia.,Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2052, Australia
| | - Celine F Santiago
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia.,Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2052, Australia
| | - Scott H Kesteven
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia.,Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2052, Australia
| | - Ze-Yan Yu
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia.,Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2052, Australia
| | - Michael P Feneley
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia.,Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2052, Australia.,Department of Cardiology, St Vincent's Hospital, Darlinghurst, New South Wales 2010, Australia
| | - Diane Fatkin
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia .,Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2052, Australia.,Department of Cardiology, St Vincent's Hospital, Darlinghurst, New South Wales 2010, Australia
| |
Collapse
|
80
|
Keen AN, Klaiman JM, Shiels HA, Gillis TE. Temperature-induced cardiac remodelling in fish. ACTA ACUST UNITED AC 2016; 220:147-160. [PMID: 27852752 PMCID: PMC5278617 DOI: 10.1242/jeb.128496] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Thermal acclimation causes the heart of some fish species to undergo significant remodelling. This includes changes in electrical activity, energy utilization and structural properties at the gross and molecular level of organization. The purpose of this Review is to summarize the current state of knowledge of temperature-induced structural remodelling in the fish ventricle across different levels of biological organization, and to examine how such changes result in the modification of the functional properties of the heart. The structural remodelling response is thought to be responsible for changes in cardiac stiffness, the Ca2+ sensitivity of force generation and the rate of force generation by the heart. Such changes to both active and passive properties help to compensate for the loss of cardiac function caused by a decrease in physiological temperature. Hence, temperature-induced cardiac remodelling is common in fish that remain active following seasonal decreases in temperature. This Review is organized around the ventricular phases of the cardiac cycle – specifically diastolic filling, isovolumic pressure generation and ejection – so that the consequences of remodelling can be fully described. We also compare the thermal acclimation-associated modifications of the fish ventricle with those seen in the mammalian ventricle in response to cardiac pathologies and exercise. Finally, we consider how the plasticity of the fish heart may be relevant to survival in a climate change context, where seasonal temperature changes could become more extreme and variable. Summary: Thermal acclimation of some temperate fishes causes extensive remodelling of the heart. The resultant changes to the active and passive properties of the heart represent a highly integrated phenotypic response.
Collapse
Affiliation(s)
- Adam N Keen
- Division of Cardiovascular Science, School of Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9NT, UK
| | - Jordan M Klaiman
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA 98109, USA
| | - Holly A Shiels
- Division of Cardiovascular Science, School of Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9NT, UK
| | - Todd E Gillis
- Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| |
Collapse
|
81
|
Marro J, Pfefferli C, de Preux Charles AS, Bise T, Jaźwińska A. Collagen XII Contributes to Epicardial and Connective Tissues in the Zebrafish Heart during Ontogenesis and Regeneration. PLoS One 2016; 11:e0165497. [PMID: 27783651 PMCID: PMC5081208 DOI: 10.1371/journal.pone.0165497] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/12/2016] [Indexed: 12/25/2022] Open
Abstract
Zebrafish heart regeneration depends on cardiac cell proliferation, epicardium activation and transient reparative tissue deposition. The contribution and the regulation of specific collagen types during the regenerative process, however, remain poorly characterized. Here, we identified that the non-fibrillar type XII collagen, which serves as a matrix-bridging component, is expressed in the epicardium of the zebrafish heart, and is boosted after cryoinjury-induced ventricular damage. During heart regeneration, an intense deposition of Collagen XII covers the outer epicardial cap and the interstitial reparative tissue. Analysis of the activated epicardium and fibroblast markers revealed a heterogeneous cellular origin of Collagen XII. Interestingly, this matrix-bridging collagen co-localized with fibrillar type I collagen and several glycoproteins in the post-injury zone, suggesting its role in tissue cohesion. Using SB431542, a selective inhibitor of the TGF-β receptor, we showed that while the inhibitor treatment did not affect the expression of collagen 12 and collagen 1a2 in the epicardium, it completely suppressed the induction of both genes in the fibrotic tissue. This suggests that distinct mechanisms might regulate collagen expression in the outer heart layer and the inner injury zone. On the basis of this study, we postulate that the TGF-β signaling pathway induces and coordinates formation of a transient collagenous network that comprises fibril-forming Collagen I and fiber-associated Collagen XII, both of which contribute to the reparative matrix of the regenerating zebrafish heart.
Collapse
Affiliation(s)
- Jan Marro
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Catherine Pfefferli
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | | | - Thomas Bise
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
- * E-mail:
| |
Collapse
|
82
|
Rubin N, Harrison MR, Krainock M, Kim R, Lien CL. Recent advancements in understanding endogenous heart regeneration-insights from adult zebrafish and neonatal mice. Semin Cell Dev Biol 2016; 58:34-40. [PMID: 27132022 PMCID: PMC5028242 DOI: 10.1016/j.semcdb.2016.04.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/13/2016] [Accepted: 04/17/2016] [Indexed: 02/06/2023]
Abstract
Enhancing the endogenous regenerative capacity of the mammalian heart is a promising strategy that can lead to potential treatment of injured cardiac tissues. Studies on heart regeneration in zebrafish and neonatal mice have shown that cardiomyocyte proliferation is essential for replenishing myocardium. We will review recent advancements that have demonstrated the importance of Neuregulin 1/ErbB2 and innervation in regulating cardiomyocyte proliferation using both adult zebrafish and neonatal mouse heart regeneration models. Emerging findings suggest that different populations of macrophages and inflammation might contribute to regenerative versus fibrotic responses. Finally, we will discuss variation in the severity of the cardiac injury and size of the wound, which may explain the range of outcomes observed in different injury models.
Collapse
Affiliation(s)
- Nicole Rubin
- Heart Institute and Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children's Hospital Los Angeles, United States; Division of Cardiothoracic Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, United States
| | - Michael R Harrison
- Heart Institute and Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children's Hospital Los Angeles, United States; Division of Cardiothoracic Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, United States
| | - Michael Krainock
- Heart Institute and Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children's Hospital Los Angeles, United States; Division of Cardiothoracic Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, United States
| | - Richard Kim
- Heart Institute and Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children's Hospital Los Angeles, United States; Division of Cardiothoracic Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, United States
| | - Ching-Ling Lien
- Heart Institute and Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children's Hospital Los Angeles, United States; Division of Cardiothoracic Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, United States; Department of Biochemistry & Molecular Biology, Keck School of Medicine, University of Southern California, United States.
| |
Collapse
|
83
|
Manalo T, May A, Quinn J, Lafontant DS, Shifatu O, He W, Gonzalez-Rosa JM, Burns GC, Burns CE, Burns AR, Lafontant PJ. Differential Lectin Binding Patterns Identify Distinct Heart Regions in Giant Danio ( Devario aequipinnatus) and Zebrafish ( Danio rerio) Hearts. J Histochem Cytochem 2016; 64:687-714. [PMID: 27680670 DOI: 10.1369/0022155416667928] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Lectins are carbohydrate-binding proteins commonly used as biochemical and histochemical tools to study glycoconjugate (glycoproteins, glycolipids) expression patterns in cells, tissues, including mammalian hearts. However, lectins have received little attention in zebrafish ( Danio rerio) and giant danio ( Devario aequipinnatus) heart studies. Here, we sought to determine the binding patterns of six commonly used lectins-wheat germ agglutinin (WGA), Ulex europaeus agglutinin, Bandeiraea simplicifolia lectin (BS lectin), concanavalin A (Con A), Ricinus communis agglutinin I (RCA I), and Lycopersicon esculentum agglutinin (tomato lectin)-in these hearts. Con A showed broad staining in the myocardium. WGA stained cardiac myocyte borders, with binding markedly stronger in the compact heart and bulbus. BS lectin, which stained giant danio coronaries, was used to measure vascular reconstruction during regeneration. However, BS lectin reacted poorly in zebrafish. RCA I stained the compact heart of both fish. Tomato lectin stained the giant danio, and while low reactivity was seen in the zebrafish ventricle, staining was observed in their transitional cardiac myocytes. In addition, we observed unique staining patterns in the developing zebrafish heart. Lectins' ability to reveal differential glycoconjugate expression in giant danio and zebrafish hearts suggests they can serve as simple but important tools in studies of developing, adult, and regenerating fish hearts.
Collapse
Affiliation(s)
- Trina Manalo
- Department of Biology, DePauw University, Greencastle, Indiana (TM, AM, JQ, DSL, OS, WH, PJL)
| | - Adam May
- Department of Biology, DePauw University, Greencastle, Indiana (TM, AM, JQ, DSL, OS, WH, PJL)
| | - Joshua Quinn
- Department of Biology, DePauw University, Greencastle, Indiana (TM, AM, JQ, DSL, OS, WH, PJL)
| | - Dominique S Lafontant
- Department of Biology, DePauw University, Greencastle, Indiana (TM, AM, JQ, DSL, OS, WH, PJL)
| | - Olubusola Shifatu
- Department of Biology, DePauw University, Greencastle, Indiana (TM, AM, JQ, DSL, OS, WH, PJL)
| | - Wei He
- Department of Biology, DePauw University, Greencastle, Indiana (TM, AM, JQ, DSL, OS, WH, PJL)
| | - Juan M Gonzalez-Rosa
- Division of Cardiology, Department of Medicine, Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, Massachusetts (JMG-R, GCB, CEB)
| | - Geoffrey C Burns
- Division of Cardiology, Department of Medicine, Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, Massachusetts (JMG-R, GCB, CEB)
| | - Caroline E Burns
- Division of Cardiology, Department of Medicine, Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, Massachusetts (JMG-R, GCB, CEB)
| | - Alan R Burns
- College of Optometry, University of Houston, Houston, Texas (ARB)
| | - Pascal J Lafontant
- Department of Biology, DePauw University, Greencastle, Indiana (TM, AM, JQ, DSL, OS, WH, PJL)
| |
Collapse
|
84
|
Cheng F, Miao L, Wu Q, Gong X, Xiong J, Zhang J. Vinculin b deficiency causes epicardial hyperplasia and coronary vessel disorganization in zebrafish. Development 2016; 143:3522-3531. [PMID: 27578788 DOI: 10.1242/dev.132936] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 07/26/2016] [Indexed: 12/29/2022]
Abstract
Coronary vessel development is a highly coordinated process during heart formation. Abnormal development and dysfunction of the coronary network are contributory factors in the majority of heart disease. Understanding the molecular mechanisms that regulate coronary vessel formation is crucial for preventing and treating the disease. We report a zebrafish gene-trap vinculin b (vclb) mutant that displays abnormal coronary vessel development among multiple cardiac defects. The mutant shows overproliferation of epicardium-derived cells and disorganization of coronary vessels, and they eventually die off at juvenile stages. Mechanistically, Vclb deficiency results in the release of another cytoskeletal protein, paxillin, from the Vclb complex and the upregulation of ERK and FAK phosphorylation in epicardium and endocardium, causing disorganization of endothelial cells and pericytes during coronary vessel development. By contrast, cardiac muscle development is relatively normal, probably owing to redundancy with Vcla, a vinculin paralog that is expressed in the myocardium but not epicardium. Together, our results reveal a previously unappreciated function of vinculin in epicardium and endocardium and reinforce the notion that well-balanced FAK activity is essential for coronary vessel development.
Collapse
Affiliation(s)
- Feng Cheng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liyun Miao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Wu
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | - Xia Gong
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jingwei Xiong
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | - Jian Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
85
|
Embryonic Ethanol Exposure Dysregulates BMP and Notch Signaling, Leading to Persistent Atrio-Ventricular Valve Defects in Zebrafish. PLoS One 2016; 11:e0161205. [PMID: 27556898 PMCID: PMC4996461 DOI: 10.1371/journal.pone.0161205] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/01/2016] [Indexed: 12/30/2022] Open
Abstract
Fetal alcohol spectrum disorder (FASD), birth defects associated with ethanol exposure in utero, includes a wide spectrum of congenital heart defects (CHDs), the most prevalent of which are septal and conotruncal defects. Zebrafish FASD model was used to dissect the mechanisms underlying FASD-associated CHDs. Embryonic ethanol exposure (3–24 hours post fertilization) led to defects in atrio-ventricular (AV) valvulogenesis beginning around 37 hpf, a morphogenetic event that arises long after ethanol withdrawal. Valve leaflets of the control embryos comprised two layers of cells confined at the compact atrio-ventricular canal (AVC). Ethanol treated embryos had extended AVC and valve forming cells were found either as rows of cells spanning the AVC or as unorganized clusters near the AV boundary. Ethanol exposure reduced valve precursors at the AVC, but some ventricular cells in ethanol treated embryos exhibited few characteristics of valve precursors. Late staged larvae and juvenile fish exposed to ethanol during embryonic development had faulty AV valves. Examination of AVC morphogenesis regulatory networks revealed that early ethanol exposure disrupted the Bmp signaling gradient in the heart during valve formation. Bmp signaling was prominent at the AVC in controls, but ethanol-exposed embryos displayed active Bmp signaling throughout the ventricle. Ethanol exposure also led to mislocalization of Notch signaling cells in endocardium during AV valve formation. Normally, highly active Notch signaling cells were organized at the AVC. In ethanol-exposed embryos, highly active Notch signaling cells were dispersed throughout the ventricle. At later stages, ethanol-exposed embryos exhibited reduced Wnt/β-catenin activity at the AVC. We conclude that early embryonic ethanol exposure alters Bmp, Notch and other signaling activities during AVC differentiation leading to faulty valve morphogenesis and valve defects persist in juvenile fish.
Collapse
|
86
|
Tahara N, Brush M, Kawakami Y. Cell migration during heart regeneration in zebrafish. Dev Dyn 2016; 245:774-87. [PMID: 27085002 PMCID: PMC5839122 DOI: 10.1002/dvdy.24411] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 03/17/2016] [Accepted: 04/12/2016] [Indexed: 12/27/2022] Open
Abstract
Zebrafish possess the remarkable ability to regenerate injured hearts as adults, which contrasts the very limited ability in mammals. Although very limited, mammalian hearts do in fact have measurable levels of cardiomyocyte regeneration. Therefore, elucidating mechanisms of zebrafish heart regeneration would provide information of naturally occurring regeneration to potentially apply to mammalian studies, in addition to addressing this biologically interesting phenomenon in itself. Studies over the past 13 years have identified processes and mechanisms of heart regeneration in zebrafish. After heart injury, pre-existing cardiomyocytes dedifferentiate, enter the cell cycle, and repair the injured myocardium. This process requires interaction with epicardial cells, endocardial cells, and vascular endothelial cells. Epicardial cells envelope the heart, while endocardial cells make up the inner lining of the heart. They provide paracrine signals to cardiomyocytes to regenerate the injured myocardium, which is vascularized during heart regeneration. In addition, accumulating results suggest that local migration of these major cardiac cell types have roles in heart regeneration. In this review, we summarize the characteristics of various heart injury methods used in the research community and regeneration of the major cardiac cell types. Then, we discuss local migration of these cardiac cell types and immune cells during heart regeneration. Developmental Dynamics 245:774-787, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Naoyuki Tahara
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - Michael Brush
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
| | - Yasuhiko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
87
|
Novel therapeutic strategies targeting fibroblasts and fibrosis in heart disease. Nat Rev Drug Discov 2016; 15:620-638. [PMID: 27339799 DOI: 10.1038/nrd.2016.89] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Our understanding of the functions of cardiac fibroblasts has moved beyond their roles in heart structure and extracellular matrix generation and now includes their contributions to paracrine, mechanical and electrical signalling during ontogenesis and normal cardiac activity. Fibroblasts also have central roles in pathogenic remodelling during myocardial ischaemia, hypertension and heart failure. As key contributors to scar formation, they are crucial for tissue repair after interventions including surgery and ablation. Novel experimental approaches targeting cardiac fibroblasts are promising potential therapies for heart disease. Indeed, several existing drugs act, at least partially, through effects on cardiac connective tissue. This Review outlines the origins and roles of fibroblasts in cardiac development, homeostasis and disease; illustrates the involvement of fibroblasts in current and emerging clinical interventions; and identifies future targets for research and development.
Collapse
|
88
|
Ernens I, Lumley AI, Devaux Y, Wagner DR. Use of Coronary Ultrasound Imaging to Evaluate Ventricular Function in Adult Zebrafish. Zebrafish 2016; 13:477-480. [PMID: 27326768 DOI: 10.1089/zeb.2016.1274] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
So far, imaging of the adult zebrafish heart and assessment of heart failure in adult zebrafish have been very limited. Here, we describe a new method for in vivo imaging of the hypertrabeculated heart of the adult zebrafish using miniaturized cardiac ultrasound catheters obtained from the cardiac catheterization laboratory. This method allows the observation of the ventricle of zebrafish and the assessment of ventricular diameters during diastole and systole, as well as heart rate and fractional shortening. Significant changes in these parameters were detected through the use of an adult zebrafish heart failure model induced by chronic anemia. This imaging technique opens the door to detailed in vivo analysis of the adult heart failure phenotype in zebrafish.
Collapse
Affiliation(s)
- Isabelle Ernens
- 1 Cardiovascular Research Unit, Luxembourg Institute of Health , Strassen, Luxembourg
| | - Andrew I Lumley
- 1 Cardiovascular Research Unit, Luxembourg Institute of Health , Strassen, Luxembourg
| | - Yvan Devaux
- 1 Cardiovascular Research Unit, Luxembourg Institute of Health , Strassen, Luxembourg
| | - Daniel R Wagner
- 1 Cardiovascular Research Unit, Luxembourg Institute of Health , Strassen, Luxembourg .,2 Division of Cardiology, Centre Hospitalier de Luxembourg , Luxembourg, Luxembourg
| |
Collapse
|
89
|
Anatomical features for the adequate choice of experimental animal models in biomedicine: I. Fishes. Ann Anat 2016; 205:75-84. [DOI: 10.1016/j.aanat.2016.02.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 12/01/2015] [Accepted: 02/01/2016] [Indexed: 11/21/2022]
|
90
|
Andrés-Delgado L, Mercader N. Interplay between cardiac function and heart development. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1707-16. [PMID: 26952935 PMCID: PMC4906158 DOI: 10.1016/j.bbamcr.2016.03.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/29/2016] [Accepted: 03/03/2016] [Indexed: 12/24/2022]
Abstract
Mechanotransduction refers to the conversion of mechanical forces into biochemical or electrical signals that initiate structural and functional remodeling in cells and tissues. The heart is a kinetic organ whose form changes considerably during development and disease. This requires cardiomyocytes to be mechanically durable and able to mount coordinated responses to a variety of environmental signals on different time scales, including cardiac pressure loading and electrical and hemodynamic forces. During physiological growth, myocytes, endocardial and epicardial cells have to adaptively remodel to these mechanical forces. Here we review some of the recent advances in the understanding of how mechanical forces influence cardiac development, with a focus on fluid flow forces. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
Affiliation(s)
- Laura Andrés-Delgado
- Development of the Epicardium and Its Role during Regeneration Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Nadia Mercader
- Development of the Epicardium and Its Role during Regeneration Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain; Institute of Anatomy, University of Bern, Bern, Switzerland.
| |
Collapse
|
91
|
Gomes RSM, Skroblin P, Munster AB, Tomlins H, Langley SR, Zampetaki A, Yin X, Wardle FC, Mayr M. "Young at heart": Regenerative potential linked to immature cardiac phenotypes. J Mol Cell Cardiol 2016; 92:105-8. [PMID: 26827899 PMCID: PMC4796039 DOI: 10.1016/j.yjmcc.2016.01.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/26/2016] [Accepted: 01/27/2016] [Indexed: 01/15/2023]
Abstract
The adult human myocardium is incapable of regeneration; yet, the zebrafish (Danio rerio) can regenerate damaged myocardium. Similar to the zebrafish heart, hearts of neonatal, but not adult mice are capable of myocardial regeneration. We performed a proteomics analysis of adult zebrafish hearts and compared their protein expression profile to hearts from neonatal and adult mice. Using difference in-gel electrophoresis (DIGE), there was little overlap between the proteome from adult mouse (> 8 weeks old) and adult zebrafish (18 months old) hearts. Similarly, there was a significant degree of mismatch between the protein expression in neonatal and adult mouse hearts. Enrichment analysis of the selected proteins revealed over-expression of DNA synthesis-related proteins in the cardiac proteome of the adult zebrafish heart similar to neonatal and 4 days old mice, whereas in hearts of adult mice there was a mitochondria-related predominance in protein expression. Importantly, we noted pronounced differences in the myofilament composition: the adult zebrafish heart lacks many of the myofilament proteins of differentiated adult cardiomyocytes such as the ventricular isoforms of myosin light chains and nebulette. Instead, troponin I and myozenin 1 were expressed as skeletal isoforms rather than cardiac isoforms. The relative immaturity of the adult zebrafish heart was further supported by cardiac microRNA data. Our assessment of zebrafish and mammalian hearts challenges the assertions on the translational potential of cardiac regeneration in the zebrafish model. The immature myofilament composition of the fish heart may explain why adult mouse and human cardiomyocytes lack this endogenous repair mechanism. Proteomics reveals minimal overlap between adult mouse and adult zebrafish hearts. Gene expression analysis confirms profound differences in myofilament composition. The adult zebrafish heart is more similar to a newborn mouse heart. The relative immaturity is further supported by cardiac microRNA data.
Collapse
Affiliation(s)
- Renata S M Gomes
- King's British Heart Foundation Centre, King's College London, London, UK
| | - Philipp Skroblin
- King's British Heart Foundation Centre, King's College London, London, UK
| | - Alex B Munster
- King's British Heart Foundation Centre, King's College London, London, UK
| | - Hannah Tomlins
- King's British Heart Foundation Centre, King's College London, London, UK
| | - Sarah R Langley
- King's British Heart Foundation Centre, King's College London, London, UK
| | - Anna Zampetaki
- King's British Heart Foundation Centre, King's College London, London, UK
| | - Xiaoke Yin
- King's British Heart Foundation Centre, King's College London, London, UK
| | - Fiona C Wardle
- Cardiovascular Development, Randall Division, King's College London, UK
| | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, London, UK.
| |
Collapse
|
92
|
Lee L, Genge CE, Cua M, Sheng X, Rayani K, Beg MF, Sarunic MV, Tibbits GF. Functional Assessment of Cardiac Responses of Adult Zebrafish (Danio rerio) to Acute and Chronic Temperature Change Using High-Resolution Echocardiography. PLoS One 2016; 11:e0145163. [PMID: 26730947 PMCID: PMC4701665 DOI: 10.1371/journal.pone.0145163] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 11/27/2015] [Indexed: 12/21/2022] Open
Abstract
The zebrafish (Danio rerio) is an important organism as a model for understanding vertebrate cardiovascular development. However, little is known about adult ZF cardiac function and how contractile function changes to cope with fluctuations in ambient temperature. The goals of this study were to: 1) determine if high resolution echocardiography (HRE) in the presence of reduced cardiodepressant anesthetics could be used to accurately investigate the structural and functional properties of the ZF heart and 2) if the effect of ambient temperature changes both acutely and chronically could be determined non-invasively using HRE in vivo. Heart rate (HR) appears to be the critical factor in modifying cardiac output (CO) with ambient temperature fluctuation as it increases from 78 ± 5.9 bpm at 18°C to 162 ± 9.7 bpm at 28°C regardless of acclimation state (cold acclimated CA- 18°C; warm acclimated WA- 28°C). Stroke volume (SV) is highest when the ambient temperature matches the acclimation temperature, though this difference did not constitute a significant effect (CA 1.17 ± 0.15 μL at 18°C vs 1.06 ± 0.14 μl at 28°C; WA 1.10 ± 0.13 μL at 18°C vs 1.12 ± 0.12 μl at 28°C). The isovolumetric contraction time (IVCT) was significantly shorter in CA fish at 18°C. The CA group showed improved systolic function at 18°C in comparison to the WA group with significant increases in both ejection fraction and fractional shortening and decreases in IVCT. The decreased early peak (E) velocity and early peak velocity / atrial peak velocity (E/A) ratio in the CA group are likely associated with increased reliance on atrial contraction for ventricular filling.
Collapse
Affiliation(s)
- Ling Lee
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
- Child and Family Research Institute, Department of Cardiovascular Science, 950 West 28th Ave, Vancouver, BC V5Z 4H4, Canada
| | - Christine E. Genge
- Child and Family Research Institute, Department of Cardiovascular Science, 950 West 28th Ave, Vancouver, BC V5Z 4H4, Canada
| | - Michelle Cua
- School of Engineering Science, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Xiaoye Sheng
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
- Child and Family Research Institute, Department of Cardiovascular Science, 950 West 28th Ave, Vancouver, BC V5Z 4H4, Canada
| | - Kaveh Rayani
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Mirza F. Beg
- School of Engineering Science, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Marinko V. Sarunic
- School of Engineering Science, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Glen F. Tibbits
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
- Child and Family Research Institute, Department of Cardiovascular Science, 950 West 28th Ave, Vancouver, BC V5Z 4H4, Canada
| |
Collapse
|
93
|
Genge CE, Lin E, Lee L, Sheng X, Rayani K, Gunawan M, Stevens CM, Li AY, Talab SS, Claydon TW, Hove-Madsen L, Tibbits GF. The Zebrafish Heart as a Model of Mammalian Cardiac Function. Rev Physiol Biochem Pharmacol 2016; 171:99-136. [PMID: 27538987 DOI: 10.1007/112_2016_5] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Zebrafish (Danio rerio) are widely used as vertebrate model in developmental genetics and functional genomics as well as in cardiac structure-function studies. The zebrafish heart has been increasingly used as a model of human cardiac function, in part, due to the similarities in heart rate and action potential duration and morphology with respect to humans. The teleostian zebrafish is in many ways a compelling model of human cardiac function due to the clarity afforded by its ease of genetic manipulation, the wealth of developmental biological information, and inherent suitability to a variety of experimental techniques. However, in addition to the numerous advantages of the zebrafish system are also caveats related to gene duplication (resulting in paralogs not present in human or other mammals) and fundamental differences in how zebrafish hearts function. In this review, we discuss the use of zebrafish as a cardiac function model through the use of techniques such as echocardiography, optical mapping, electrocardiography, molecular investigations of excitation-contraction coupling, and their physiological implications relative to that of the human heart. While some of these techniques (e.g., echocardiography) are particularly challenging in the zebrafish because of diminutive size of the heart (~1.5 mm in diameter) critical information can be derived from these approaches and are discussed in detail in this article.
Collapse
Affiliation(s)
- Christine E Genge
- Molecular Cardiac Physiology Group, Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada, V5A 1S6
| | - Eric Lin
- Molecular Cardiac Physiology Group, Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada, V5A 1S6
| | - Ling Lee
- BC Children's Hospital Research Institute, Vancouver, BC, Canada, V5Z 4H4
| | - XiaoYe Sheng
- BC Children's Hospital Research Institute, Vancouver, BC, Canada, V5Z 4H4
| | - Kaveh Rayani
- Molecular Cardiac Physiology Group, Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada, V5A 1S6
| | - Marvin Gunawan
- Molecular Cardiac Physiology Group, Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada, V5A 1S6
| | - Charles M Stevens
- Molecular Cardiac Physiology Group, Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada, V5A 1S6.,BC Children's Hospital Research Institute, Vancouver, BC, Canada, V5Z 4H4
| | - Alison Yueh Li
- Molecular Cardiac Physiology Group, Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada, V5A 1S6
| | - Sanam Shafaat Talab
- Molecular Cardiac Physiology Group, Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada, V5A 1S6
| | - Thomas W Claydon
- Molecular Cardiac Physiology Group, Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada, V5A 1S6
| | - Leif Hove-Madsen
- Molecular Cardiac Physiology Group, Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada, V5A 1S6.,Cardiovascular Research Centre CSIC-ICCC, Hospital de Sant Pau, Barcelona, Spain
| | - Glen F Tibbits
- Molecular Cardiac Physiology Group, Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada, V5A 1S6. .,BC Children's Hospital Research Institute, Vancouver, BC, Canada, V5Z 4H4.
| |
Collapse
|
94
|
Jaźwińska A, Sallin P. Regeneration versus scarring in vertebrate appendages and heart. J Pathol 2016; 238:233-46. [PMID: 26414617 PMCID: PMC5057359 DOI: 10.1002/path.4644] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/15/2015] [Accepted: 09/18/2015] [Indexed: 12/15/2022]
Abstract
Injuries to complex human organs, such as the limbs and the heart, result in pathological conditions, for which we often lack adequate treatments. While modern regenerative approaches are based on the transplantation of stem cell-derived cells, natural regeneration in lower vertebrates, such as zebrafish and newts, relies predominantly on the intrinsic plasticity of mature tissues. This property involves local activation of the remaining material at the site of injury to promote cell division, cell migration and complete reproduction of the missing structure. It remains an unresolved question why adult mammals are not equally competent to reactivate morphogenetic programmes. Although organ regeneration depends strongly on the proliferative properties of cells in the injured tissue, it is apparent that various organismic factors, such as innervation, vascularization, hormones, metabolism and the immune system, can affect this process. Here, we focus on a correlation between the regenerative capacity and cellular specialization in the context of functional demands, as illustrated by appendages and heart in diverse vertebrates. Elucidation of the differences between homologous regenerative and non-regenerative tissues from various animal models is essential for understanding the applicability of lessons learned from the study of regenerative biology to clinical strategies for the treatment of injured human organs.
Collapse
Affiliation(s)
- Anna Jaźwińska
- Department of Biology, University of Fribourg, Switzerland
| | - Pauline Sallin
- Department of Biology, University of Fribourg, Switzerland
| |
Collapse
|
95
|
Hemodynamics driven cardiac valve morphogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1760-6. [PMID: 26608609 DOI: 10.1016/j.bbamcr.2015.11.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/13/2015] [Accepted: 11/17/2015] [Indexed: 11/22/2022]
Abstract
Mechanical forces are instrumental to cardiovascular development and physiology. The heart beats approximately 2.6 billion times in a human lifetime and heart valves ensure that these contractions result in an efficient, unidirectional flow of the blood. Composed of endocardial cells (EdCs) and extracellular matrix (ECM), cardiac valves are among the most mechanically challenged structures of the body both during and after their development. Understanding how hemodynamic forces modulate cardiovascular function and morphogenesis is key to unraveling the relationship between normal and pathological cardiovascular development and physiology. Most valve diseases have their origins in embryogenesis, either as signs of abnormal developmental processes or the aberrant re-expression of fetal gene programs normally quiescent in adulthood. Here we review recent discoveries in the mechanobiology of cardiac valve development and introduce the latest technologies being developed in the zebrafish, including live cell imaging and optical technologies, as well as modeling approaches that are currently transforming this field. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
|
96
|
Lee J, Cao H, Kang BJ, Jen N, Yu F, Lee CA, Fei P, Park J, Bohlool S, Lash-Rosenberg L, Shung KK, Hsiai TK. Hemodynamics and ventricular function in a zebrafish model of injury and repair. Zebrafish 2015; 11:447-54. [PMID: 25237983 DOI: 10.1089/zeb.2014.1016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Myocardial infarction results in scar tissue and irreversible loss of ventricular function. Unlike humans, zebrafish has the capacity to remove scar tissue after injury. To assess ventricular function during repair, we synchronized microelectrocardiogram (μECG) signals with a high-frequency ultrasound pulsed-wave (PW) Doppler to interrogate cardiac hemodynamics. μECG signals allowed for identification of PW Doppler signals for passive (early [E]-wave velocity) and active ventricular filling (atrial [A]-wave velocity) during diastole. The A wave (9.0±1.2 cm·s(-1)) is greater than the E wave (1.1±0.4 cm·s(-1)), resulting in an E/A ratio <1 (0.12±0.05, n=6). In response to cryocauterization to the ventricular epicardium, the E-wave velocity increased, accompanied by a rise in the E/A ratio at 3 days postcryocauterization (dpc) (0.55±0.13, n=6, p<0.001 vs. sham). The E waves normalize toward the baseline, along with a reduction in the E/A ratio at 35 dpc (0.36±0.06, n=6, p<0.001 vs. sham) and 65 dpc (0.2±0.16, n=6, p<0.001 vs. sham). In zebrafish, E/A<1 at baseline is observed, suggesting the distinct two-chamber system in which the pressure gradient across the atrioventricular valve is higher compared with the ventriculobulbar valve. The initial rise and subsequent normalization of E/A ratios support recovery in the ventricular diastolic function.
Collapse
Affiliation(s)
- Juhyun Lee
- 1 Division of Cardiology, Department of Medicine, University of California , Los Angeles, Los Angeles, California
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Leone M, Magadum A, Engel FB. Cardiomyocyte proliferation in cardiac development and regeneration: a guide to methodologies and interpretations. Am J Physiol Heart Circ Physiol 2015; 309:H1237-50. [PMID: 26342071 DOI: 10.1152/ajpheart.00559.2015] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The newt and the zebrafish have the ability to regenerate many of their tissues and organs including the heart. Thus, a major goal in experimental medicine is to elucidate the molecular mechanisms underlying the regenerative capacity of these species. A wide variety of experiments have demonstrated that naturally occurring heart regeneration relies on cardiomyocyte proliferation. Thus, major efforts have been invested to induce proliferation of mammalian cardiomyocytes in order to improve cardiac function after injury or to protect the heart from further functional deterioration. In this review, we describe and analyze methods currently used to evaluate cardiomyocyte proliferation. In addition, we summarize the literature on naturally occurring heart regeneration. Our analysis highlights that newt and zebrafish heart regeneration relies on factors that are also utilized in cardiomyocyte proliferation during mammalian fetal development. Most of these factors have, however, failed to induce adult mammalian cardiomyocyte proliferation. Finally, our analysis of mammalian neonatal heart regeneration indicates experiments that could resolve conflicting results in the literature, such as binucleation assays and clonal analysis. Collectively, cardiac regeneration based on cardiomyocyte proliferation is a promising approach for improving adult human cardiac function after injury, but it is important to elucidate the mechanisms arresting mammalian cardiomyocyte proliferation after birth and to utilize better assays to determine formation of new muscle mass.
Collapse
Affiliation(s)
- Marina Leone
- Experimental Renal and Cardiovascular Research, Institute of Pathology, Department of Nephropathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany; and
| | - Ajit Magadum
- Department of Cardiology, Icahn School of Medicine at Mount Sinai Hospital, New York, New York
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Institute of Pathology, Department of Nephropathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany; and
| |
Collapse
|
98
|
van Soldt BJ, Danielsen CC, Wang T. The mechanical properties of the systemic and pulmonary arteries of Python regius correlate with blood pressures. J Morphol 2015; 276:1412-21. [PMID: 26780263 DOI: 10.1002/jmor.20429] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 07/02/2015] [Accepted: 07/03/2015] [Indexed: 11/07/2022]
Abstract
Pythons are unique amongst snakes in having different pressures in the aortas and pulmonary arteries because of intraventricular pressure separation. In this study, we investigate whether this correlates with different blood vessel strength in the ball python Python regius. We excised segments from the left, right, and dorsal aortas, and from the two pulmonary arteries. These were subjected to tensile testing. We show that the aortic vessel wall is significantly stronger than the pulmonary artery wall in P. regius. Gross morphological characteristics (vessel wall thickness and correlated absolute amount of collagen content) are likely the most influential factors. Collagen fiber thickness and orientation are likely to have an effect, though the effect of collagen fiber type and cross-links between fibers will need further study.
Collapse
Affiliation(s)
- Benjamin J van Soldt
- Department of Bioscience, Zoophysiology, University of Aarhus, C.F. Møllers Allé 3, Aarhus C, 8000, Denmark
- Department of Genetics and Development, Columbia University Medical Center, New York, New York
| | | | - Tobias Wang
- Department of Bioscience, Zoophysiology, University of Aarhus, C.F. Møllers Allé 3, Aarhus C, 8000, Denmark
| |
Collapse
|
99
|
Cooper TK, Spitsbergen JM. Valvular and Mural Endocardiosis in Aging Zebrafish (Danio rerio). Vet Pathol 2015; 53:504-9. [DOI: 10.1177/0300985815594853] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Endocardiosis or myxomatous degeneration of the cardiac valves is a well-described age-related change in humans and dogs. Lesions consist of polypoid nodular proliferations of loose extracellular matrix and valvular interstitial cells, most commonly affecting the mitral valve. This entity has not been previously described in fish. Herein we report the appearance, location, and occurrence of valvular and mural endocardiosis in a retrospective survey of aging laboratory zebrafish. Endocardiosis was present in 59 of 777 fish (7.59%), most commonly affecting the sinoatrial (34 fish; 57.6%) and atrioventricular (33 fish; 55.9%) valves. Lesions were more common in fish raised in recirculating water systems and fed commercial diets (52/230 fish; 22.6%) versus flow-through systems with fish fed semi-purified diets (4/234; 1.71%). Lesions were overrepresented in fish heterozygous for a mutant smoothened allele (34/61 fish, 55.7% vs 17/168, 10.1% wild type). There was no association between endocardiosis and intestinal carcinoids. Valvular endocardiosis is a significant age- and husbandry-related background finding in zebrafish and should be considered in the design and interpretation of research studies.
Collapse
Affiliation(s)
- T. K. Cooper
- Penn State Hershey Medical Center, Departments of Comparative Medicine and Pathology, Hershey, PA, USA
| | | |
Collapse
|
100
|
Mersereau EJ, Poitra SL, Espinoza A, Crossley DA, Darland T. The effects of cocaine on heart rate and electrocardiogram in zebrafish (Danio rerio). Comp Biochem Physiol C Toxicol Pharmacol 2015; 172-173:1-6. [PMID: 25847362 PMCID: PMC4458413 DOI: 10.1016/j.cbpc.2015.03.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/25/2015] [Accepted: 03/27/2015] [Indexed: 10/23/2022]
Abstract
Zebrafish (Danio rerio) have been used as a model organism to explore the genetic basis for responsiveness to addictive drugs like cocaine. However, very little is known about how the physiological response to cocaine is mediated in zebrafish. In the present study electrocardiograms (ECGs) were recorded from adult zebrafish treated with cocaine. Treatment with cocaine resulted in a bell-shaped dose response curve with a maximal change in heart rate seen using 5mg/L cocaine. Higher doses resulted in a higher percentage of fish showing bradycardia. The cocaine-induced tachycardia was blocked by co-treatment with propranolol, a β-adrenergic antagonist, but potentiated by co-treatment with phentolamine, an α-adrenergic antagonist. Co-treatment with atropine, a classic cholinergic antagonist, had no effect on cocaine-induced tachycardia. Cocaine treatment of adult fish changed the ECG of treated fish, inducing a dose-dependent increase in QT interval after adjusting for heart rate (QTc), while not affecting the PR or QRS intervals. The acute effects of cocaine on heart rate were examined in 5-day old embryos to see if zebrafish might serve as a suitable model organism to study possible links of embryonic physiological response to subsequent adult behavioral response to the drug. Cocaine treatment of 5-day old zebrafish embryos also resulted in a bell-shaped dose response curve, with maximal tachycardia achieved with 10mg/L. The response in embryonic fish was thus comparable to that in adults and raises the possibility that the effects of embryonic exposure to cocaine on the developing cardiovascular system can be effectively modeled in zebrafish.
Collapse
Affiliation(s)
- Erik J Mersereau
- University of North Dakota, Biology Department, Grand Forks, ND 58202-9019, USA
| | - Shelby L Poitra
- University of North Dakota, Biology Department, Grand Forks, ND 58202-9019, USA
| | - Ana Espinoza
- University of Arizona, Department of Ecology and Evolutionary Biology, Tucson, AZ 85721, USA
| | - Dane A Crossley
- University of North Texas, Department of Biological Sciences, Denton, TX 76203, USA
| | - Tristan Darland
- University of North Dakota, Biology Department, Grand Forks, ND 58202-9019, USA.
| |
Collapse
|