151
|
Parisi C, Vashisht S, Winata CL. Fish-Ing for Enhancers in the Heart. Int J Mol Sci 2021; 22:3914. [PMID: 33920121 PMCID: PMC8069060 DOI: 10.3390/ijms22083914] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/19/2022] Open
Abstract
Precise control of gene expression is crucial to ensure proper development and biological functioning of an organism. Enhancers are non-coding DNA elements which play an essential role in regulating gene expression. They contain specific sequence motifs serving as binding sites for transcription factors which interact with the basal transcription machinery at their target genes. Heart development is regulated by intricate gene regulatory network ensuring precise spatiotemporal gene expression program. Mutations affecting enhancers have been shown to result in devastating forms of congenital heart defect. Therefore, identifying enhancers implicated in heart biology and understanding their mechanism is key to improve diagnosis and therapeutic options. Despite their crucial role, enhancers are poorly studied, mainly due to a lack of reliable way to identify them and determine their function. Nevertheless, recent technological advances have allowed rapid progress in enhancer discovery. Model organisms such as the zebrafish have contributed significant insights into the genetics of heart development through enabling functional analyses of genes and their regulatory elements in vivo. Here, we summarize the current state of knowledge on heart enhancers gained through studies in model organisms, discuss various approaches to discover and study their function, and finally suggest methods that could further advance research in this field.
Collapse
Affiliation(s)
- Costantino Parisi
- International Institute of Molecular and Cell Biology in Warsaw, 02-109 Warsaw, Poland; (C.P.); (S.V.)
| | - Shikha Vashisht
- International Institute of Molecular and Cell Biology in Warsaw, 02-109 Warsaw, Poland; (C.P.); (S.V.)
| | - Cecilia Lanny Winata
- International Institute of Molecular and Cell Biology in Warsaw, 02-109 Warsaw, Poland; (C.P.); (S.V.)
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| |
Collapse
|
152
|
Persistent Ventricle Partitioning in the Adult Zebrafish Heart. J Cardiovasc Dev Dis 2021; 8:jcdd8040041. [PMID: 33918756 PMCID: PMC8070482 DOI: 10.3390/jcdd8040041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 02/07/2023] Open
Abstract
The vertebrate heart integrates cells from the early-differentiating first heart field (FHF) and the later-differentiating second heart field (SHF), both emerging from the lateral plate mesoderm. In mammals, this process forms the basis for the development of the left and right ventricle chambers and subsequent chamber septation. The single ventricle-forming zebrafish heart also integrates FHF and SHF lineages during embryogenesis, yet the contributions of these two myocardial lineages to the adult zebrafish heart remain incompletely understood. Here, we characterize the myocardial labeling of FHF descendants in both the developing and adult zebrafish ventricle. Expanding previous findings, late gastrulation-stage labeling using drl-driven CreERT2 recombinase with a myocardium-specific, myl7-controlled, loxP reporter results in the predominant labeling of FHF-derived outer curvature and the right side of the embryonic ventricle. Raised to adulthood, such lineage-labeled hearts retain broad areas of FHF cardiomyocytes in a region of the ventricle that is positioned at the opposite side to the atrium and encompasses the apex. Our data add to the increasing evidence for a persisting cell-based compartmentalization of the adult zebrafish ventricle even in the absence of any physical boundary.
Collapse
|
153
|
Zebrafish Models of Autosomal Recessive Ataxias. Cells 2021; 10:cells10040836. [PMID: 33917666 PMCID: PMC8068028 DOI: 10.3390/cells10040836] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022] Open
Abstract
Autosomal recessive ataxias are much less well studied than autosomal dominant ataxias and there are no clearly defined systems to classify them. Autosomal recessive ataxias, which are characterized by neuronal and multisystemic features, have significant overlapping symptoms with other complex multisystemic recessive disorders. The generation of animal models of neurodegenerative disorders increases our knowledge of their cellular and molecular mechanisms and helps in the search for new therapies. Among animal models, the zebrafish, which shares 70% of its genome with humans, offer the advantages of being small in size and demonstrating rapid development, making them optimal for high throughput drug and genetic screening. Furthermore, embryo and larval transparency allows to visualize cellular processes and central nervous system development in vivo. In this review, we discuss the contributions of zebrafish models to the study of autosomal recessive ataxias characteristic phenotypes, behavior, and gene function, in addition to commenting on possible treatments found in these models. Most of the zebrafish models generated to date recapitulate the main features of recessive ataxias.
Collapse
|
154
|
Majumdar U, Yasuhara J, Garg V. In Vivo and In Vitro Genetic Models of Congenital Heart Disease. Cold Spring Harb Perspect Biol 2021; 13:cshperspect.a036764. [PMID: 31818859 DOI: 10.1101/cshperspect.a036764] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Congenital cardiovascular malformations represent the most common type of birth defect and encompass a spectrum of anomalies that range from mild to severe. The etiology of congenital heart disease (CHD) is becoming increasingly defined based on prior epidemiologic studies that supported the importance of genetic contributors and technological advances in human genome analysis. These have led to the discovery of a growing number of disease-contributing genetic abnormalities in individuals affected by CHD. The ever-growing population of adult CHD survivors, which are the result of reductions in mortality from CHD during childhood, and this newfound genetic knowledge have led to important questions regarding recurrence risks, the mechanisms by which these defects occur, the potential for novel approaches for prevention, and the prediction of long-term cardiovascular morbidity in adult CHD survivors. Here, we will review the current status of genetic models that accurately model human CHD as they provide an important tool to answer these questions and test novel therapeutic strategies.
Collapse
Affiliation(s)
- Uddalak Majumdar
- Center for Cardiovascular Research, Nationwide Children's Hospital, Columbus, Ohio 43205, USA.,The Heart Center, Nationwide Children's Hospital, Columbus, Ohio 43205, USA
| | - Jun Yasuhara
- Center for Cardiovascular Research, Nationwide Children's Hospital, Columbus, Ohio 43205, USA.,The Heart Center, Nationwide Children's Hospital, Columbus, Ohio 43205, USA
| | - Vidu Garg
- Center for Cardiovascular Research, Nationwide Children's Hospital, Columbus, Ohio 43205, USA.,The Heart Center, Nationwide Children's Hospital, Columbus, Ohio 43205, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio 43205, USA.,Department of Molecular Genetics, The Ohio State University, Columbus, Ohio 43205, USA
| |
Collapse
|
155
|
Ma J, Huang Y, Peng Y, Xu Z, Wang Z, Chen X, Xie S, Jiang P, Zhong K, Lu H. Bifenazate exposure induces cardiotoxicity in zebrafish embryos. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 274:116539. [PMID: 33549839 DOI: 10.1016/j.envpol.2021.116539] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/24/2020] [Accepted: 01/18/2021] [Indexed: 06/12/2023]
Abstract
Bifenazate is a novel acaricide for selective foliar spraying and is widely used to control mites in agricultural production. However, its toxicity to aquatic organisms is unknown. Here, a zebrafish model was used to study bifenazate toxicity to aquatic organisms. Exposure to bifenazate was found to cause severe cardiotoxicity in zebrafish embryos, along with disorders in the gene expression related to heart development. Bifenazate also caused oxidative stress. Cardiotoxicity caused by bifenazate was partially rescued by astaxanthin (an antioxidant), accompanied by cardiac genes and oxidative stress-related indicators becoming normalized. Our results showed that exposure to bifenazate can significantly change the ATPase activity and gene expression levels of the calcium signaling pathway. These led to heart failure, in which the blood accumulated outside the heart without entering it, eventually leading to death. The results indicated that bifenazate exposure caused cardiotoxicity in zebrafish embryos through the induction of oxidative stress and inhibition of the calcium signaling pathway.
Collapse
Affiliation(s)
- Jinze Ma
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Yong Huang
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000 Jiangxi, China
| | - Yuyang Peng
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Zhaopeng Xu
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Ziqin Wang
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Xiaobei Chen
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Shuling Xie
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Ping Jiang
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Keyuan Zhong
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Huiqiang Lu
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000 Jiangxi, China; Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, Affiliated Hospital of Jinggangshan University, Ji'an, 343009, China.
| |
Collapse
|
156
|
Rufaihah AJ, Chen CK, Yap CH, Mattar CNZ. Mending a broken heart: In vitro, in vivo and in silico models of congenital heart disease. Dis Model Mech 2021; 14:dmm047522. [PMID: 33787508 PMCID: PMC8033415 DOI: 10.1242/dmm.047522] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Birth defects contribute to ∼0.3% of global infant mortality in the first month of life, and congenital heart disease (CHD) is the most common birth defect among newborns worldwide. Despite the significant impact on human health, most treatments available for this heterogenous group of disorders are palliative at best. For this reason, the complex process of cardiogenesis, governed by multiple interlinked and dose-dependent pathways, is well investigated. Tissue, animal and, more recently, computerized models of the developing heart have facilitated important discoveries that are helping us to understand the genetic, epigenetic and mechanobiological contributors to CHD aetiology. In this Review, we discuss the strengths and limitations of different models of normal and abnormal cardiogenesis, ranging from single-cell systems and 3D cardiac organoids, to small and large animals and organ-level computational models. These investigative tools have revealed a diversity of pathogenic mechanisms that contribute to CHD, including genetic pathways, epigenetic regulators and shear wall stresses, paving the way for new strategies for screening and non-surgical treatment of CHD. As we discuss in this Review, one of the most-valuable advances in recent years has been the creation of highly personalized platforms with which to study individual diseases in clinically relevant settings.
Collapse
Affiliation(s)
- Abdul Jalil Rufaihah
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
| | - Ching Kit Chen
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
| | - Choon Hwai Yap
- Division of Cardiology, Department of Paediatrics, Khoo Teck Puat -National University Children's Medical Institute, National University Health System, Singapore 119228
- Department of Bioengineering, Imperial College London, London, UK
| | - Citra N Z Mattar
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
- Department of Obstetrics and Gynaecology, National University Health System, Singapore 119228
| |
Collapse
|
157
|
Kemmler CL, Riemslagh FW, Moran HR, Mosimann C. From Stripes to a Beating Heart: Early Cardiac Development in Zebrafish. J Cardiovasc Dev Dis 2021; 8:17. [PMID: 33578943 PMCID: PMC7916704 DOI: 10.3390/jcdd8020017] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 12/18/2022] Open
Abstract
The heart is the first functional organ to form during vertebrate development. Congenital heart defects are the most common type of human birth defect, many originating as anomalies in early heart development. The zebrafish model provides an accessible vertebrate system to study early heart morphogenesis and to gain new insights into the mechanisms of congenital disease. Although composed of only two chambers compared with the four-chambered mammalian heart, the zebrafish heart integrates the core processes and cellular lineages central to cardiac development across vertebrates. The rapid, translucent development of zebrafish is amenable to in vivo imaging and genetic lineage tracing techniques, providing versatile tools to study heart field migration and myocardial progenitor addition and differentiation. Combining transgenic reporters with rapid genome engineering via CRISPR-Cas9 allows for functional testing of candidate genes associated with congenital heart defects and the discovery of molecular causes leading to observed phenotypes. Here, we summarize key insights gained through zebrafish studies into the early patterning of uncommitted lateral plate mesoderm into cardiac progenitors and their regulation. We review the central genetic mechanisms, available tools, and approaches for modeling congenital heart anomalies in the zebrafish as a representative vertebrate model.
Collapse
Affiliation(s)
| | | | | | - Christian Mosimann
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine and Children’s Hospital Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (C.L.K.); (F.W.R.); (H.R.M.)
| |
Collapse
|
158
|
Martin KE, Waxman JS. Atrial and Sinoatrial Node Development in the Zebrafish Heart. J Cardiovasc Dev Dis 2021; 8:jcdd8020015. [PMID: 33572147 PMCID: PMC7914448 DOI: 10.3390/jcdd8020015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/31/2021] [Accepted: 02/04/2021] [Indexed: 12/11/2022] Open
Abstract
Proper development and function of the vertebrate heart is vital for embryonic and postnatal life. Many congenital heart defects in humans are associated with disruption of genes that direct the formation or maintenance of atrial and pacemaker cardiomyocytes at the venous pole of the heart. Zebrafish are an outstanding model for studying vertebrate cardiogenesis, due to the conservation of molecular mechanisms underlying early heart development, external development, and ease of genetic manipulation. Here, we discuss early developmental mechanisms that instruct appropriate formation of the venous pole in zebrafish embryos. We primarily focus on signals that determine atrial chamber size and the specialized pacemaker cells of the sinoatrial node through directing proper specification and differentiation, as well as contemporary insights into the plasticity and maintenance of cardiomyocyte identity in embryonic zebrafish hearts. Finally, we integrate how these insights into zebrafish cardiogenesis can serve as models for human atrial defects and arrhythmias.
Collapse
Affiliation(s)
- Kendall E. Martin
- Molecular Genetics, Biochemistry, and Microbiology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Joshua S. Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Correspondence:
| |
Collapse
|
159
|
Mousavi SE, Purser GJ, Patil JG. Embryonic Onset of Sexually Dimorphic Heart Rates in the Viviparous Fish, Gambusia holbrooki. Biomedicines 2021; 9:165. [PMID: 33567532 PMCID: PMC7915484 DOI: 10.3390/biomedicines9020165] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
In fish, little is known about sex-specific differences in physiology and performance of the heart and whether these differences manifest during development. Here for the first time, the sex-specific heart rates during embryogenesis of Gambusia holbrooki, from the onset of the heart rates (HRs) to just prior to parturition, was investigated using light cardiogram. The genetic sex of the embryos was post-verified using a sex-specific genetic marker. Results reveal that heart rates and resting time significantly increase (p < 0.05) with progressive embryonic development. Furthermore, both ventricular and atrial frequencies of female embryos were significantly higher (p < 0.05) than those of their male sibs at the corresponding developmental stages and remained so at all later developmental stages (p < 0.05). In concurrence, the heart rate and ventricular size of the adult females were also significantly (p < 0.05) higher and larger respectively than those of males. Collectively, the results suggest that the cardiac sex-dimorphism manifests as early as late-organogenesis and persists through adulthood in this species. These findings suggest that the cardiac measurements can be employed to non-invasively sex the developing embryos, well in advance of when their phenotypic sex is discernible. In addition, G. holbrooki could serve as a better model to study comparative vertebrate cardiovascular development as well as to investigate anthropogenic and climatic impacts on heart physiology of this species, that may be sex influenced.
Collapse
Affiliation(s)
- Seyed Ehsan Mousavi
- Fisheries and Aquaculture Centre, Institute for Marine and Antarctic Studies, University of Tasmania, Taroona, TAS 7053, Australia;
| | - G. John Purser
- Fisheries and Aquaculture Centre, Institute for Marine and Antarctic Studies, University of Tasmania, Taroona, TAS 7053, Australia;
| | - Jawahar G. Patil
- Fisheries and Aquaculture Centre, Institute for Marine and Antarctic Studies, University of Tasmania, Taroona, TAS 7053, Australia;
- Inland Fisheries Service, New Norfolk, TAS 7140, Australia
| |
Collapse
|
160
|
Wan-Mohtar WAAQI, Ilham Z, Jamaludin AA, Rowan N. Use of Zebrafish Embryo Assay to Evaluate Toxicity and Safety of Bioreactor-Grown Exopolysaccharides and Endopolysaccharides from European Ganoderma applanatum Mycelium for Future Aquaculture Applications. Int J Mol Sci 2021; 22:1675. [PMID: 33562361 PMCID: PMC7914815 DOI: 10.3390/ijms22041675] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 01/30/2021] [Accepted: 02/04/2021] [Indexed: 12/15/2022] Open
Abstract
Natural mycelial exopolysaccharide (EPS) and endopolysaccharide (ENS) extracted from bioreactor-cultivated European Ganoderma applanatum mushrooms are of potential high commercial value for both food and adjacent biopharmaceutical industries. In order to evaluate their potential toxicity for aquaculture application, both EPS (0.01-10 mg/mL) and ENS (0.01-10 mg/mL) extracts were tested for Zebrafish Embryo Toxicity (ZFET); early development effects on Zebrafish Embryos (ZE) were also analyzed between 24 and 120 h post-fertilization (HPF). Both EPS and ENS are considered non-toxic with LC50 of 1.41 mg/mL and 0.87 mg/mL respectively. Both EPS and ENS did not delay hatching and teratogenic defect towards ZE with <1.0 mg/mL, respectively. No significant changes in the ZE heart rate were detected following treatment with the two compounds tested (EPS: 0.01-10 mg/mL: 176.44 ± 0.77 beats/min and ENS: 0.01-10 mg/mL: 148.44 ± 17.75 beats/min) compared to normal ZE (120-180 beats/min). These initial findings support future pre-clinical trials in adult fish models with view to safely using EPS and ENS as potential feed supplements for supplements for development of the aquaculture industry.
Collapse
Affiliation(s)
- Wan Abd Al Qadr Imad Wan-Mohtar
- Functional Omics and Bioprocess Development Laboratory, Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
- Bioresources and Bioprocessing Research Group, Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
- Bioscience Research Institute, Athlone Institute of Technology, Dublin Road, N37 WO89 Athlone, Westmeath, Ireland
| | - Zul Ilham
- Bioresources and Bioprocessing Research Group, Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
- Environmental Science and Management Program, Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Adi Ainurzaman Jamaludin
- Environmental Science and Management Program, Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Neil Rowan
- Bioscience Research Institute, Athlone Institute of Technology, Dublin Road, N37 WO89 Athlone, Westmeath, Ireland
- Empower Eco Innovation Hub, Lough Boora, Co., R35 DA50 Tullamore, Offaly, Ireland
| |
Collapse
|
161
|
Han CR, Wang H, Hoffmann V, Zerfas P, Kruhlak M, Cheng SY. Thyroid Hormone Receptor α Mutations Cause Heart Defects in Zebrafish. Thyroid 2021; 31:315-326. [PMID: 32762296 PMCID: PMC7891307 DOI: 10.1089/thy.2020.0332] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background: Mutations of thyroid hormone receptor α1 (TRα1) cause resistance to thyroid hormone (RTHα). Patients exhibit growth retardation, delayed bone development, anemia, and bradycardia. By using mouse models of RTHα, much has been learned about the molecular actions of TRα1 mutants that underlie these abnormalities in adults. Using zebrafish models of RTHα that we have recently created, we aimed to understand how TRα1 mutants affect the heart function during this period. Methods: In contrast to human and mice, the thra gene is duplicated, thraa and thrab, in zebrafish. Using CRISPR/Cas9-mediated targeted mutagenesis, we created C-terminal mutations in each of two duplicated thra genes in zebrafish (thraa 8-bp insertion or thrab 1-bp insertion mutations). We recently showed that these mutant fish faithfully recapitulated growth retardation as found in patients and thra mutant mice. In the present study, we used histological analysis, gene expression profiles, confocal fluorescence, and transmission electron microscopy (TEM) to comprehensively analyze the phenotypic characteristics of mutant fish heart during development. Results: We found both a dilated atrium and an abnormally shaped ventricle in adult mutant fish. The retention of red blood cells in the two abnormal heart chambers, and the decreased circulating blood speed and reduced expression of contractile genes indicated weakened contractility in the heart of mutant fish. These abnormalities were detected in mutant fish as early as 35 days postfertilization (juveniles). Furthermore, the expression of genes associated with the sarcomere assembly was suppressed in the heart of mutant fish, resulting in abnormalities of sarcomere organization as revealed by TEM, suggesting that the abnormal sarcomere organization could underlie the bradycardia exhibited in mutant fish. Conclusions: Using a zebrafish model of RTHα, the present study demonstrated for the first time that TRα1 mutants could act to cause abnormal heart structure, weaken contractility, and disrupt sarcomere organization that affect heart functions. These findings provide new insights into the bradycardia found in RTHα patients.
Collapse
Affiliation(s)
- Cho Rong Han
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Hui Wang
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Victoria Hoffmann
- Diagnostic and Research Services Branch, Office of Research Services, National Institutes of Health, Bethesda, Maryland, USA
| | - Patricia Zerfas
- Diagnostic and Research Services Branch, Office of Research Services, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael Kruhlak
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Sheue-Yann Cheng
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
- Address correspondence to: Sheue-Yann Cheng, PhD, Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 5128, Bethesda, MD 20892-4264, USA
| |
Collapse
|
162
|
Lane S, More LA, Asnani A. Zebrafish Models of Cancer Therapy-Induced Cardiovascular Toxicity. J Cardiovasc Dev Dis 2021; 8:jcdd8020008. [PMID: 33499052 PMCID: PMC7911266 DOI: 10.3390/jcdd8020008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/11/2021] [Accepted: 01/20/2021] [Indexed: 12/13/2022] Open
Abstract
Purpose of review: Both traditional and novel cancer therapies can cause cardiovascular toxicity in patients. In vivo models integrating both cardiovascular and cancer phenotypes allow for the study of on- and off-target mechanisms of toxicity arising from these agents. The zebrafish is the optimal whole organism model to screen for cardiotoxicity in a high throughput manner, while simultaneously assessing the role of cardiotoxicity pathways on the cancer therapy’s antitumor effect. Here we highlight established zebrafish models of human cardiovascular disease and cancer, the unique advantages of zebrafish to study mechanisms of cancer therapy-associated cardiovascular toxicity, and finally, important limitations to consider when using the zebrafish to study toxicity. Recent findings: Cancer therapy-associated cardiovascular toxicities range from cardiomyopathy with traditional agents to arrhythmias and thrombotic complications associated with newer targeted therapies. The zebrafish can be used to identify novel therapeutic strategies that selectively protect the heart from cancer therapy without affecting antitumor activity. Advances in genome editing technology have enabled the creation of several transgenic zebrafish lines valuable to the study of cardiovascular and cancer pathophysiology. Summary: The high degree of genetic conservation between zebrafish and humans, as well as the ability to recapitulate cardiotoxic phenotypes observed in patients with cancer, make the zebrafish an effective model to study cancer therapy-associated cardiovascular toxicity. Though this model provides several key benefits over existing in vitro and in vivo models, limitations of the zebrafish model include the early developmental stage required for most high-throughput applications.
Collapse
Affiliation(s)
- Sarah Lane
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.L.); (L.A.M.)
| | - Luis Alberto More
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.L.); (L.A.M.)
| | - Aarti Asnani
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.L.); (L.A.M.)
- Harvard Medical School, Boston, MA 02115, USA
- Correspondence:
| |
Collapse
|
163
|
Filice M, Barca A, Amelio D, Leo S, Mazzei A, Del Vecchio G, Verri T, Cerra MC, Imbrogno S. Morpho-functional remodelling of the adult zebrafish (Danio rerio) heart in response to waterborne angiotensin II exposure. Gen Comp Endocrinol 2021; 301:113663. [PMID: 33220301 DOI: 10.1016/j.ygcen.2020.113663] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/04/2020] [Accepted: 11/12/2020] [Indexed: 12/29/2022]
Abstract
Angiotensin II (AngII), the principal effector of the Renin-Angiotensin System, is a pluripotent humoral agent whose biological actions include short-term modulations and long-term adaptations. In fish, short-term cardio-tropic effects of AngII are documented, but information on the role of AngII in long-term cardiac remodelling is not fully understood. Here, we describe a direct approach to disclose long-term morpho-functional effects of AngII on the zebrafish heart. Adult fish exposed to waterborne teleost analogue AngII for 8 weeks showed enhanced heart weight and cardio-somatic index, coupled to myocardial structural changes (i.e. augmented compacta thickness and fibrosis), and increased heart rate. These findings were paralleled by an up-regulation of type-1 and type-2 AngII receptors expression, and by changes in the expression of GATA binding protein 4, nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 and superoxide dismutase 1 soluble mRNAs, as well as of cytochrome b-245 beta polypeptide protein, indicative of cardiac remodelling. Our results suggest that waterborne AngII can sustain and robustly affect the cardiac morpho-functional remodelling of adult zebrafish.
Collapse
Affiliation(s)
- Mariacristina Filice
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende, CS, Italy
| | - Amilcare Barca
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni, I-73100 Lecce, Italy
| | - Daniela Amelio
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende, CS, Italy
| | - Serena Leo
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende, CS, Italy
| | - Aurora Mazzei
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni, I-73100 Lecce, Italy
| | - Gianmarco Del Vecchio
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni, I-73100 Lecce, Italy
| | - Tiziano Verri
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni, I-73100 Lecce, Italy
| | - Maria Carmela Cerra
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende, CS, Italy
| | - Sandra Imbrogno
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende, CS, Italy.
| |
Collapse
|
164
|
Simpson KE, Venkateshappa R, Pang ZK, Faizi S, Tibbits GF, Claydon TW. Utility of Zebrafish Models of Acquired and Inherited Long QT Syndrome. Front Physiol 2021; 11:624129. [PMID: 33519527 PMCID: PMC7844309 DOI: 10.3389/fphys.2020.624129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/21/2020] [Indexed: 01/12/2023] Open
Abstract
Long-QT Syndrome (LQTS) is a cardiac electrical disorder, distinguished by irregular heart rates and sudden death. Accounting for ∼40% of cases, LQTS Type 2 (LQTS2), is caused by defects in the Kv11.1 (hERG) potassium channel that is critical for cardiac repolarization. Drug block of hERG channels or dysfunctional channel variants can result in acquired or inherited LQTS2, respectively, which are typified by delayed repolarization and predisposition to lethal arrhythmia. As such, there is significant interest in clear identification of drugs and channel variants that produce clinically meaningful perturbation of hERG channel function. While toxicological screening of hERG channels, and phenotypic assessment of inherited channel variants in heterologous systems is now commonplace, affordable, efficient, and insightful whole organ models for acquired and inherited LQTS2 are lacking. Recent work has shown that zebrafish provide a viable in vivo or whole organ model of cardiac electrophysiology. Characterization of cardiac ion currents and toxicological screening work in intact embryos, as well as adult whole hearts, has demonstrated the utility of the zebrafish model to contribute to the development of therapeutics that lack hERG-blocking off-target effects. Moreover, forward and reverse genetic approaches show zebrafish as a tractable model in which LQTS2 can be studied. With the development of new tools and technologies, zebrafish lines carrying precise channel variants associated with LQTS2 have recently begun to be generated and explored. In this review, we discuss the present knowledge and questions raised related to the use of zebrafish as models of acquired and inherited LQTS2. We focus discussion, in particular, on developments in precise gene-editing approaches in zebrafish to create whole heart inherited LQTS2 models and evidence that zebrafish hearts can be used to study arrhythmogenicity and to identify potential anti-arrhythmic compounds.
Collapse
Affiliation(s)
- Kyle E. Simpson
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Ravichandra Venkateshappa
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Zhao Kai Pang
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Shoaib Faizi
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Glen F. Tibbits
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- Department of Cardiovascular Science, British Columbia Children’s Hospital, Vancouver, BC, Canada
| | - Tom W. Claydon
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
165
|
Echeazarra L, Hortigón-Vinagre MP, Casis O, Gallego M. Adult and Developing Zebrafish as Suitable Models for Cardiac Electrophysiology and Pathology in Research and Industry. Front Physiol 2021; 11:607860. [PMID: 33519514 PMCID: PMC7838705 DOI: 10.3389/fphys.2020.607860] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022] Open
Abstract
The electrophysiological behavior of the zebrafish heart is very similar to that of the human heart. In fact, most of the genes that codify the channels and regulatory proteins required for human cardiac function have their orthologs in the zebrafish. The high fecundity, small size, and easy handling make the zebrafish embryos/larvae an interesting candidate to perform whole animal experiments within a plate, offering a reliable and low-cost alternative to replace rodents and larger mammals for the study of cardiac physiology and pathology. The employment of zebrafish embryos/larvae has widened from basic science to industry, being of particular interest for pharmacology studies, since the zebrafish embryo/larva is able to recapitulate a complete and integrated view of cardiac physiology, missed in cell culture. As in the human heart, IKr is the dominant repolarizing current and it is functional as early as 48 h post fertilization. Finally, genome editing techniques such as CRISPR/Cas9 facilitate the humanization of zebrafish embryos/larvae. These techniques allow one to replace zebrafish genes by their human orthologs, making humanized zebrafish embryos/larvae the most promising in vitro model, since it allows the recreation of human-organ-like environment, which is especially necessary in cardiac studies due to the implication of dynamic factors, electrical communication, and the paracrine signals in cardiac function.
Collapse
Affiliation(s)
- Leyre Echeazarra
- Departamento de Fisiología, Facultad de Farmacia, Universidad del País Vasco UPV/EHU, Vitoria-Gasteiz, Spain
| | - Maria Pura Hortigón-Vinagre
- Departamento de Bioquímica y Biología Molecular y Genética>, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Oscar Casis
- Departamento de Fisiología, Facultad de Farmacia, Universidad del País Vasco UPV/EHU, Vitoria-Gasteiz, Spain
| | - Mónica Gallego
- Departamento de Fisiología, Facultad de Farmacia, Universidad del País Vasco UPV/EHU, Vitoria-Gasteiz, Spain
| |
Collapse
|
166
|
The effect of ginsenoside Rg5, isolated from black ginseng, on heart failure in zebrafish based on untargeted metabolomics. J Funct Foods 2021. [DOI: 10.1016/j.jff.2020.104325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
167
|
Carlantoni C, Allanki S, Kontarakis Z, Rossi A, Piesker J, Günther S, Stainier DY. Tie1 regulates zebrafish cardiac morphogenesis through Tolloid-like 1 expression. Dev Biol 2021; 469:54-67. [DOI: 10.1016/j.ydbio.2020.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/10/2020] [Accepted: 09/13/2020] [Indexed: 01/13/2023]
|
168
|
van de Venter M, Didloff J, Reddy S, Swanepoel B, Govender S, Dambuza NS, Williams S, Koekemoer TC, Venables L. Wild-Type Zebrafish ( Danio rerio) Larvae as a Vertebrate Model for Diabetes and Comorbidities: A Review. Animals (Basel) 2020; 11:E54. [PMID: 33396883 PMCID: PMC7824285 DOI: 10.3390/ani11010054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022] Open
Abstract
Zebrafish have become a popular alternative to higher animals in biomedical and pharmaceutical research. The development of stable mutant lines to model target specific aspects of many diseases, including diabetes, is well reported. However, these mutant lines are much more costly and challenging to maintain than wild-type zebrafish and are simply not an option for many research facilities. As an alternative to address the disadvantages of advanced mutant lines, wild-type larvae may represent a suitable option. In this review, we evaluate organ development in zebrafish larvae and discuss established methods that use wild-type zebrafish larvae up to seven days post fertilization to test for potential drug candidates for diabetes and its commonly associated conditions of oxidative stress and inflammation. This provides an up to date overview of the relevance of wild-type zebrafish larvae as a vertebrate antidiabetic model and confidence as an alternative tool for preclinical studies. We highlight the advantages and disadvantages of established methods and suggest recommendations for future developments to promote the use of zebrafish, specifically larvae, rather than higher animals in the early phase of antidiabetic drug discovery.
Collapse
Affiliation(s)
- Maryna van de Venter
- Department of Biochemistry and Microbiology, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa; (J.D.); (S.R.); (B.S.); (S.G.); (S.W.); (T.C.K.); (L.V.)
| | - Jenske Didloff
- Department of Biochemistry and Microbiology, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa; (J.D.); (S.R.); (B.S.); (S.G.); (S.W.); (T.C.K.); (L.V.)
| | - Shanika Reddy
- Department of Biochemistry and Microbiology, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa; (J.D.); (S.R.); (B.S.); (S.G.); (S.W.); (T.C.K.); (L.V.)
| | - Bresler Swanepoel
- Department of Biochemistry and Microbiology, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa; (J.D.); (S.R.); (B.S.); (S.G.); (S.W.); (T.C.K.); (L.V.)
| | - Sharlene Govender
- Department of Biochemistry and Microbiology, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa; (J.D.); (S.R.); (B.S.); (S.G.); (S.W.); (T.C.K.); (L.V.)
| | - Ntokozo Shirley Dambuza
- Department of Pharmacy, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa;
| | - Saralene Williams
- Department of Biochemistry and Microbiology, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa; (J.D.); (S.R.); (B.S.); (S.G.); (S.W.); (T.C.K.); (L.V.)
| | - Trevor Craig Koekemoer
- Department of Biochemistry and Microbiology, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa; (J.D.); (S.R.); (B.S.); (S.G.); (S.W.); (T.C.K.); (L.V.)
| | - Luanne Venables
- Department of Biochemistry and Microbiology, Nelson Mandela University, PO Box 77000, Port Elizabeth 6031, South Africa; (J.D.); (S.R.); (B.S.); (S.G.); (S.W.); (T.C.K.); (L.V.)
| |
Collapse
|
169
|
Huang Y, Chen Z, Meng Y, Wei Y, Xu Z, Ma J, Zhong K, Cao Z, Liao X, Lu H. Famoxadone-cymoxanil induced cardiotoxicity in zebrafish embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 205:111339. [PMID: 32961491 DOI: 10.1016/j.ecoenv.2020.111339] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 06/11/2023]
Abstract
Famoxadone-cymoxanil is a new protective and therapeutic fungicide, but little research has been done on it or its toxicity in aquatic organisms. In this study, we used zebrafish to investigate the cardiotoxicity of famoxadone-cymoxanil and the potential mechanisms involved. Zebrafish embryos were exposed to different concentrations of famoxadone-cymoxanil until 72 h post-fertilization (hpf), then changes of heart morphology in zebrafish embryos were observed. We also detected the levels of oxidative stress, myocardial-cell proliferation and apoptosis, ATPase activity, and the expression of genes related to the cardiac development and calcium-signaling pathway. After famoxadone-cymoxanil exposure, pericardial edema, cardiac linearization, and reductions in the heart rate and cardiac output positively correlated with concentration. Although myocardial-cell apoptosis was not detected, proliferation of the cells was severely reduced and ATPase activity significantly decreased, resulting in a severe deficiency in heart function. In addition, indicators of oxidative stress changed significantly after exposure of the embryos to the fungicide. To better understand the possible molecular mechanisms of cardiovascular toxicity in zebrafish, we studied the transcriptional levels of cardiac development, calcium-signaling pathways, and genes associated with myocardial contractility. The mRNA expression levels of key genes in heart development were significantly down-regulated, while the expression of genes related to the calcium-signaling pathway (ATPase [atp2a1], cardiac troponin C [tnnc1a], and calcium channel [cacna1a]) was significantly inhibited. Expression of klf2a, a major endocardial flow-responsive gene, was also significantly inhibited. Mechanistically, famoxadone-cymoxanil toxicity might be due to the downregulation of genes associated with the calcium-signaling pathway and cardiac muscle contraction. Our results found that famoxadone-cymoxanil exposure causes cardiac developmental toxicity and severe energy deficiency in zebrafish.
Collapse
Affiliation(s)
- Yong Huang
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Zhiyong Chen
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, 343009, Jiangxi, China
| | - Yunlong Meng
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - You Wei
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Zhaopeng Xu
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Jinze Ma
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Keyuan Zhong
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Zigang Cao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, 343009, Jiangxi, China
| | - Xinjun Liao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, 343009, Jiangxi, China
| | - Huiqiang Lu
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, 343009, Jiangxi, China.
| |
Collapse
|
170
|
Rajesh V, Deepan N, Anitha V, Kalaiselvan D, Jayaseelan S, Sivakumar P, Ganesan V. Heart malformation is an early response to valproic acid in developing zebrafish. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:2387-2409. [PMID: 32699959 DOI: 10.1007/s00210-020-01949-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 07/13/2020] [Indexed: 10/23/2022]
Abstract
Valproic acid (VPA) is a branched short-chain fatty acid primarily used in epilepsy, but is also used in bipolar disorder, migraine, and psychotic disorders. Despite its wide range of use, it is a teratogen resulting in various congenital abnormalities. Although a large number of scientific studies evidenced the teratogenic effects, there are limited data on embryonic exposure to VPA at specific or different stages of early embryogenesis. Based on this, the present study was planned to investigate the embryonic exposure to VPA at specific and different hours post fertilization (hpf) in zebrafish embryonic model. In first set of experiments, embryos from spawning groups of adult zebrafish were exposed to different molar concentrations of VPA at 2.5 hpf, and in the second set of experiments, embryos were exposed to VPA 100 μM at 24 hpf, 36 hpf, 48 hpf, 72 hpf, and 96 hpf. The parameters examined were hatching rate, mortality, morphology, body length, pericardial sac size, heartrate, anatomical changes in heart, skeletal and notochord till 120 hpf. It was observed that the embryos exposed to VPA at 2.5 hpf suffered from cardiac abnormalities including heart malformation, bradycardia, circulatory failure, and pericardial sac enlargement which was more apparent in embryos exposed to 100 μM VPA. In the second set of experiments, embryos exposed to VPA 100 μM at 24 hpf and 36 hpf suffered from heart malformations, but there was no incidence of cardiac malformation in embryos exposed to VPA at 48 hpf, 72 hpf, and 96 hpf. From the results, it was evident that exposure to VPA at early developmental stage of embryogenesis produced congenital cardiac abnormalities. Since VPA is a selective HDAC inhibitor, histone acetylation with aberrant gene expression during cardiogenesis might be the underlying cause of cardiac malformation.
Collapse
Affiliation(s)
- Venugopalan Rajesh
- Department of Pharmacology, The Erode College of Pharmacy, Veppampalayam, Vallipurathampalayam (Po), Erode, Tamil Nadu, 638112, India.
| | - Natarajan Deepan
- Department of Pharmacology, The Erode College of Pharmacy, Veppampalayam, Vallipurathampalayam (Po), Erode, Tamil Nadu, 638112, India
| | - Vijayakumar Anitha
- Department of Pharmacology, The Erode College of Pharmacy, Veppampalayam, Vallipurathampalayam (Po), Erode, Tamil Nadu, 638112, India
| | - Duraisamy Kalaiselvan
- Department of Pharmacology, The Erode College of Pharmacy, Veppampalayam, Vallipurathampalayam (Po), Erode, Tamil Nadu, 638112, India
| | - Subramanian Jayaseelan
- Department of Pharmaceutical Analysis, The Erode College of Pharmacy, Veppampalayam, Vallipurathampalayam (Po), Erode, Tamil Nadu, 638112, India
| | - Palanivel Sivakumar
- Department of Pharmaceutical Chemistry, The Erode College of Pharmacy, Veppampalayam, Vallipurathampalayam (Po), Erode, Tamil Nadu, 638112, India
| | - Vellaiyachamy Ganesan
- Department of Pharmaceutics, The Erode College of Pharmacy, Veppampalayam, Vallipurathampalayam (Po), Erode, Tamil Nadu, 638112, India
| |
Collapse
|
171
|
Liu J, Liu Y, Yu H, Zhang Y, Hsu ACY, Zhang M, Gou Y, Sun W, Wang F, Li P, Liu J. Design, synthesis and biological evaluation of novel pyxinol derivatives with anti-heart failure activity. Biomed Pharmacother 2020; 133:111050. [PMID: 33378957 DOI: 10.1016/j.biopha.2020.111050] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/15/2020] [Accepted: 11/19/2020] [Indexed: 01/20/2023] Open
Abstract
Heart failure (HF) is an important and leading cause of substantial morbidity and mortality globally. The angiotensin-converting enzymatic (ACE) is the causative source for congestive heart failure. Natural products and its derivatives play a vital role in drug discovery and development owing to their efficacy and low toxicity. Pyxinol is a potent natural agent for cardiovascular disease. Thus we investigated the effect on ACE and HF of pyxinol derivatives. We designed and synthesized 32 novel fatty acid ester derivatives of pyxinol via esterification. Among them, compounds 2e (IC50=105 nM) and 3b (IC50=114 nM) displayed excellent ACE inhibitory activity in vitro, and exhibited non-toxic to H9c2 cells. The interactions between ACE and compounds were predicted by molecular docking respectively. In verapamil-induced zebrafish HF model, the activity assay showed that these two derivatives could improve cardiovascular physiological indexes including heart beats, venous congestion, heart dilation, cardiac output, ejection fraction and fractional shortening in a dose-dependent manner. A UPLC-QTOF-MS-based serum metabolomics approach was applied to explore the latent mechanism. A total of 25 differentiated metabolites and 8 perturbed metabolic pathways were identified. These results indicated that pyxinol fatty acid ester derivatives 2e and 3b might be considered as potent drug candidates against heart failure and deserved further research and development.
Collapse
Affiliation(s)
- Junli Liu
- School of Pharmaceutical Sciences, Jilin University, Fujin Road 1266, Changchun 130021, China
| | - Yunhe Liu
- School of Pharmaceutical Sciences, Jilin University, Fujin Road 1266, Changchun 130021, China
| | - Hui Yu
- School of Pharmaceutical Sciences, Jilin University, Fujin Road 1266, Changchun 130021, China
| | - Ying Zhang
- School of Pharmaceutical Sciences, Jilin University, Fujin Road 1266, Changchun 130021, China; The First Hospital of Jilin University, Changchun 130021, China
| | - Alan Chen-Yu Hsu
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW 2305, Australia
| | - Mingming Zhang
- College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yawei Gou
- College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Wei Sun
- College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Fang Wang
- College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Pingya Li
- School of Pharmaceutical Sciences, Jilin University, Fujin Road 1266, Changchun 130021, China
| | - Jinping Liu
- School of Pharmaceutical Sciences, Jilin University, Fujin Road 1266, Changchun 130021, China.
| |
Collapse
|
172
|
Mef2c factors are required for early but not late addition of cardiomyocytes to the ventricle. Dev Biol 2020; 470:95-107. [PMID: 33245870 PMCID: PMC7819464 DOI: 10.1016/j.ydbio.2020.11.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 11/15/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022]
Abstract
During heart formation, the heart grows and undergoes dramatic morphogenesis to achieve efficient embryonic function. Both in fish and amniotes, much of the growth occurring after initial heart tube formation arises from second heart field (SHF)-derived progenitor cell addition to the arterial pole, allowing chamber formation. In zebrafish, this process has been extensively studied during embryonic life, but it is unclear how larval cardiac growth occurs beyond 3 days post-fertilisation (dpf). By quantifying zebrafish myocardial growth using live imaging of GFP-labelled myocardium we show that the heart grows extensively between 3 and 5 dpf. Using methods to assess cell division, cellular development timing assay and Kaede photoconversion, we demonstrate that proliferation, CM addition, and hypertrophy contribute to ventricle growth. Mechanistically, we show that reduction in Mef2c activity (mef2ca+/-;mef2cb-/-), downstream or in parallel with Nkx2.5 and upstream of Ltbp3, prevents some CM addition and differentiation, resulting in a significantly smaller ventricle by 3 dpf. After 3 dpf, however, CM addition in mef2ca+/-;mef2cb-/- mutants recovers to a normal pace, and the heart size gap between mutants and their siblings diminishes into adulthood. Thus, as in mice, there is an early time window when SHF contribution to the myocardium is particularly sensitive to loss of Mef2c activity.
Collapse
|
173
|
Ding Y, Bu H, Xu X. Modeling Inherited Cardiomyopathies in Adult Zebrafish for Precision Medicine. Front Physiol 2020; 11:599244. [PMID: 33329049 PMCID: PMC7717946 DOI: 10.3389/fphys.2020.599244] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/30/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiomyopathies are a highly heterogeneous group of heart muscle disorders. More than 100 causative genes have been linked to various cardiomyopathies, which explain about half of familial cardiomyopathy cases. More than a dozen candidate therapeutic signaling pathways have been identified; however, precision medicine is not being used to treat the various types of cardiomyopathy because knowledge is lacking for how to tailor treatment plans for different genetic causes. Adult zebrafish (Danio rerio) have a higher throughout than rodents and are an emerging vertebrate model for studying cardiomyopathy. Herein, we review progress in the past decade that has proven the feasibility of this simple vertebrate for modeling inherited cardiomyopathies of distinct etiology, identifying effective therapeutic strategies for a particular type of cardiomyopathy, and discovering new cardiomyopathy genes or new therapeutic strategies via a forward genetic approach. On the basis of this progress, we discuss future research that would benefit from integrating this emerging model, including discovery of remaining causative genes and development of genotype-based therapies. Studies using this efficient vertebrate model are anticipated to significantly accelerate the implementation of precision medicine for inherited cardiomyopathies.
Collapse
Affiliation(s)
- Yonghe Ding
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Haisong Bu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States.,Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
174
|
Machikhin AS, Volkov MV, Burlakov AB, Khokhlov DD, Potemkin AV. Blood Vessel Imaging at Pre-Larval Stages of Zebrafish Embryonic Development. Diagnostics (Basel) 2020; 10:diagnostics10110886. [PMID: 33143148 PMCID: PMC7692510 DOI: 10.3390/diagnostics10110886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 01/25/2023] Open
Abstract
The zebrafish (Danio rerio) is an increasingly popular animal model biological system. In cardiovascular research, it has been used to model specific cardiac phenomena as well as to identify novel therapies for human cardiovascular disease. While the zebrafish cardiovascular system functioning is well examined at larval stages, the mechanisms by which vessel activity is initiated remain a subject of intense investigation. In this research, we report on an in vivo stain-free blood vessel imaging technique at pre-larval stages of zebrafish embryonic development. We have developed the algorithm for the enhancement, alignment and spatiotemporal analysis of bright-field microscopy images of zebrafish embryos. It enables the detection, mapping and quantitative characterization of cardiac activity across the whole specimen. To validate the proposed approach, we have analyzed multiple data cubes, calculated vessel images and evaluated blood flow velocity and heart rate dynamics in the absence of any anesthesia. This non-invasive technique may shed light on the mechanism of vessel activity initiation and stabilization as well as the cardiovascular system’s susceptibility to environmental stressors at early developmental stages.
Collapse
Affiliation(s)
- Alexander S. Machikhin
- Laboratory of Acousto-optical Spectroscopy, Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, 117342 Moscow, Russia;
| | - Mikhail V. Volkov
- Department of Applied Optics, University ITMO, 190000 Saint Petersburg, Russia; (M.V.V.); (A.V.P.)
| | - Alexander B. Burlakov
- Department of Ichthyology, Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Demid D. Khokhlov
- Laboratory of Acousto-optical Spectroscopy, Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, 117342 Moscow, Russia;
- Correspondence:
| | - Andrey V. Potemkin
- Department of Applied Optics, University ITMO, 190000 Saint Petersburg, Russia; (M.V.V.); (A.V.P.)
| |
Collapse
|
175
|
Jensen B, Christoffels VM, Moorman AFM. An Appreciation of Anatomy in the Molecular World. J Cardiovasc Dev Dis 2020; 7:E44. [PMID: 33076272 PMCID: PMC7712948 DOI: 10.3390/jcdd7040044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 12/29/2022] Open
Abstract
Robert H. Anderson is one of the most important and accomplished cardiac anatomists of the last decades, having made major contributions to our understanding of the anatomy of normal hearts and the pathologies of acquired and congenital heart diseases. While cardiac anatomy as a research discipline has become largely subservient to molecular biology, anatomists like Professor Anderson demonstrate anatomy has much to offer. Here, we provide cases of early anatomical insights on the heart that were rediscovered, and expanded on, by molecular techniques: migration of neural crest cells to the heart was deduced from histological observations (1908) and independently shown again with experimental interventions; pharyngeal mesoderm is added to the embryonic heart (1973) in what is now defined as the molecularly distinguishable second heart field; chambers develop from the heart tube as regional pouches in what is now considered the ballooning model by the molecular identification of regional differentiation and proliferation. The anatomical discovery of the conduction system by Purkinje, His, Tawara, Keith, and Flack is a special case because the main findings were never neglected in later molecular studies. Professor Anderson has successfully demonstrated that sound knowledge of anatomy is indispensable for proper understanding of cardiac development.
Collapse
Affiliation(s)
- Bjarke Jensen
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Meibergdreef 15, 1105AZ Amsterdam, The Netherlands; (V.M.C.); (A.F.M.M.)
| | | | | |
Collapse
|
176
|
Martini D, Pucci C, Gabellini C, Pellegrino M, Andreazzoli M. Exposure to the natural alkaloid Berberine affects cardiovascular system morphogenesis and functionality during zebrafish development. Sci Rep 2020; 10:17358. [PMID: 33060638 PMCID: PMC7566475 DOI: 10.1038/s41598-020-73661-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 09/17/2020] [Indexed: 12/16/2022] Open
Abstract
The plant-derived natural alkaloid berberine displays therapeutic potential to treat several pathological conditions, including dyslipidemias, diabetes and cardiovascular disorders. However, data on berberine effects during embryonic development are scarce and in part controversial. In this study, using zebrafish embryos as vertebrate experimental model, we address the effects of berberine treatment on cardiovascular system development and functionality. Starting from the observation that berberine induces developmental toxicity and pericardial edema in a time- and concentration-dependent manner, we found that treated embryos display cardiac looping defects and, at later stages, present an abnormal heart characterized by a stretched morphology and atrial endocardial/myocardial detachment. Furthermore, berberine affected cardiac functionality of the embryos, promoting bradycardia and reducing the cardiac output, the atrial shortening fraction percentage and the atrial stroke volume. We also found that, during development, berberine interferes with the angiogenic process, without altering vascular permeability. These alterations are associated with increased levels of vascular endothelial growth factor aa (vegfaa) mRNA, suggesting an important role for Vegfaa as mediator of berberine-induced cardiovascular defects. Altogether, these data indicate that berberine treatment during vertebrate development leads to an impairment of cardiovascular system morphogenesis and functionality, suggesting a note of caution in its use during pregnancy and lactation.
Collapse
Affiliation(s)
- Davide Martini
- Cell and Developmental Biology Unit, Department of Biology, University of Pisa, SS12 Abetone e Brennero, 56127, Pisa, Italy
| | - Cecilia Pucci
- Cell and Developmental Biology Unit, Department of Biology, University of Pisa, SS12 Abetone e Brennero, 56127, Pisa, Italy.,Sant'Anna School of Advanced Studies, Pisa, Italy.,Institute of Genomic Medicine, Catholic University, 00168, Rome, Italy
| | - Chiara Gabellini
- Cell and Developmental Biology Unit, Department of Biology, University of Pisa, SS12 Abetone e Brennero, 56127, Pisa, Italy
| | - Mario Pellegrino
- National Institute of Optics, National Research Council, Pisa, Italy
| | - Massimiliano Andreazzoli
- Cell and Developmental Biology Unit, Department of Biology, University of Pisa, SS12 Abetone e Brennero, 56127, Pisa, Italy. .,Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa, Italy.
| |
Collapse
|
177
|
Zebrafish as a Successful Animal Model for Screening Toxicity of Medicinal Plants. PLANTS 2020; 9:plants9101345. [PMID: 33053800 PMCID: PMC7601530 DOI: 10.3390/plants9101345] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/07/2020] [Accepted: 10/07/2020] [Indexed: 12/15/2022]
Abstract
The zebrafish (Danio rerio) is used as an embryonic and larval model to perform in vitro experiments and developmental toxicity studies. Zebrafish may be used to determine the toxicity of samples in early screening assays, often in a high-throughput manner. The zebrafish embryotoxicity model is at the leading edge of toxicology research due to the short time required for analyses, transparency of embryos, short life cycle, high fertility, and genetic data similarity. Zebrafish toxicity studies range from assessing the toxicity of bioactive compounds or crude extracts from plants to determining the optimal process. Most of the studied extracts were polar, such as ethanol, methanol, and aqueous solutions, which were used to detect the toxicity and bioactivity. This review examines the latest research using zebrafish as a study model and highlights its power as a tool for detecting toxicity of medicinal plants and its effectiveness at enhancing the understanding of new drug generation. The goal of this review was to develop a link to ethnopharmacological zebrafish studies that can be used by other researchers to conduct future research.
Collapse
|
178
|
Shen Y, Lu H, Chen R, Zhu L, Song G. MicroRNA-29c affects zebrafish cardiac development via targeting Wnt4. Mol Med Rep 2020; 22:4675-4684. [PMID: 33173954 PMCID: PMC7646856 DOI: 10.3892/mmr.2020.11584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 09/18/2020] [Indexed: 01/07/2023] Open
Abstract
As a single cardiac malformation, ventricular septal defect (VSD) is the most common form of congenital heart disease. However, the precise molecular mechanisms underlying VSD are not completely understood. Numerous microRNAs (miRs/miRNAs) are associated with ventricular septal defects. miR-29c inhibits the proliferation and promotes the apoptosis and differentiation of P19 embryonal carcinoma cells, possibly via suppressing Wnt4 signaling. However, to the best of our knowledge, no in vivo studies have been published to determine whether overexpression of miR-29c leads to developmental abnormalities. The present study was designed to observe the effect of miRNA-29c on cardiac development and its possible mechanism in vivo. Zebrafish embryos were microinjected with different doses (1, 1.6 and 2 µmol) miR-29c mimics or negative controls, and hatchability, mortality and cardiac malformation were subsequently observed. The results showed that in zebrafish embryos, miR-29c overexpression attenuated heart development in a dose-dependent manner, manifested by heart rate slowdown, pericardial edema and heart looping disorder. Further experiments showed that overexpression of miR-29c was associated with the Wnt4/β-catenin signaling pathway to regulate zebrafish embryonic heart development. In conclusion, the present results demonstrated that miR-29c regulated the lateral development and cardiac circulation of zebrafish embryo by targeting Wnt4.
Collapse
Affiliation(s)
- Yahui Shen
- Department of Respiratory and Critical Care Medicine, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Huiyu Lu
- Department of Respiratory and Critical Care Medicine, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Rong Chen
- Department of Respiratory and Critical Care Medicine, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Li Zhu
- Department of Cardiology, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Guixian Song
- Department of Cardiology, Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| |
Collapse
|
179
|
Wang D, Zhang Y, Li J, Dahlgren RA, Wang X, Huang H, Wang H. Risk assessment of cardiotoxicity to zebrafish (Danio rerio) by environmental exposure to triclosan and its derivatives. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 265:114995. [PMID: 32554097 DOI: 10.1016/j.envpol.2020.114995] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/02/2020] [Accepted: 06/06/2020] [Indexed: 06/11/2023]
Abstract
Triclosan (TCS) and its two derivatives (2,4-dichlorophenol and 2,4,6-trichlorophenol) are priority pollutants that coexist in aquatic environments. Joint exposure of TCS, 2,4-dichlorophenol and 2,4,6-trichlorophenol, hereafter referred to as TCS-DT, contributes severe toxicity to aquatic organisms. There is currently a paucity of data regarding TCS-DT molecular toxicity, especially on cardiac diseases. We used zebrafish (Danio rerio) as a model organism, and evaluated the molecular-level cardiotoxicity induced by TCS-DT from embryonic to adult stages. TCS-DT exposure prominently led to phenotypic malformations, such as pericardial cysts, cardiac bleeding, increased SV-BA distance, decreased heart rate and reduced ejection fraction, as well as abnormal swimming behavior. Analyses of the GO and KEGG pathways revealed enrichment pathways related to cardiac development and screened for significantly down-regulated adrenaline signaling in cardiomyocytes. The cardiac marker genes (amhc, cmlc2, vmhc, and nkx2.5) were obtained through protein-protein interaction (PPI) networks, and expressed as down-regulation by WISH. After chronic exposure to TCS-DT from 30 to 90-dpf, both body mass and heart indexes prominently increased, showing myocardial hypertrophy, abnormal heart rate and histopathological injury. Heart tissue damage included disordered and ruptured myocardial fibers, broken and dissolved myofilaments, nuclear pyknosis, mitochondrial injury and inflammatory cell infiltration. Further, abnormal changes in a series of cardiac functions-related biomarkers, including superoxide dismutase, triglyceride, lactate dehydrogenase and creatinine kinase MB, provided evidence for cardiac pathological responses. These results highlight the molecular mechanisms involving TCS-DT induced cardiac toxicity, and provide theoretical data to guide prevention and treatment of pollutant-induced cardiac diseases.
Collapse
Affiliation(s)
- Danting Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yuhuan Zhang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jieyi Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Randy A Dahlgren
- Department of Land, Air and Water Resources, University of California, Davis, CA, 95616, USA
| | - Xuedong Wang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, Jiangsu, 215009, China
| | - Haishan Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Huili Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
180
|
Mohankumar T, Lalithamba HS, Manigandan K, Muthaiyan A, Elangovan N. DHF-BAHPC molecule exerts ameliorative antioxidant status and reduced cadmium-induced toxicity in zebrafish (Danio rerio) embryos. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 79:103425. [PMID: 32470610 DOI: 10.1016/j.etap.2020.103425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 04/04/2020] [Accepted: 05/24/2020] [Indexed: 06/11/2023]
Abstract
In this study, we report the antioxidant and antitoxic potential of chemically synthesized 4-oxo-2-phenyl-4H-chromene-7,8-diyl bis((1-amino-2-hydroxypropyl)carbamate) (DHF-BAHPC) compound using in vitro and in vivo assays. The DHF-BAHPC was synthesized by linking 7,8-Dihydroxyflavone (DHF) with two molecules of Fmoc-threonine and characterized by Ultraviolet-visible spectroscopy (UV-vis), Fourier-transform infrared spectroscopy (FT-IR), 1H NMR, 13C NMR and Mass spectrometry (MS). In vitro, antioxidant assay results revealed that DHF-BAHPC has a dose-dependent radical scavenging potential towards DPPH, ABTS, FRAP and H2O2 radicals with an IC50 range of 15.45, 66.27, 25.71, 4.375 μg/mL, respectively. Furthermore DHF-BAHPC treatment significantly altered cadmium (Cd) intoxicated zebrafish embryos by rescuing the developmental changes associated with severe histological and reduced the level of defensive antioxidant activities (SOD, CAT, GPx and GST). The overall results of the present study represented that DHF-BAHPC may be used as a potential drug in redox-based therapeutics.
Collapse
Affiliation(s)
- Thangavel Mohankumar
- Department of Biotechnology, School of Biosciences, Periyar University, Salem 636011, Tamil Nadu, India.
| | | | - Krishnan Manigandan
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea.
| | - Arunachalam Muthaiyan
- Division of Mathematics, Physical and Natural Sciences, University of New Mexico, Gallup, NM 87301, USA.
| | - Namasivayam Elangovan
- Department of Biotechnology, School of Biosciences, Periyar University, Salem 636011, Tamil Nadu, India.
| |
Collapse
|
181
|
Huang Y, Ma J, Meng Y, Wei Y, Xie S, Jiang P, Wang Z, Chen X, Liu Z, Zhong K, Cao Z, Liao X, Xiao J, Lu H. Exposure to Oxadiazon-Butachlor causes cardiac toxicity in zebrafish embryos. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 265:114775. [PMID: 32504889 DOI: 10.1016/j.envpol.2020.114775] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/30/2020] [Accepted: 05/07/2020] [Indexed: 06/11/2023]
Abstract
Oxadiazon-Butachlor (OB) is a widely used herbicide for controlling most annual weeds in rice fields. However, its potential toxicity in aquatic organisms has not been evaluated so far. We used the zebrafish embryo model to assess the toxicity of OB, and found that it affected early cardiac development and caused extensive cardiac damage. Mechanistically, OB significantly increased oxidative stress in the embryos by inhibiting antioxidant enzymes that resulted in excessive production of reactive oxygen species (ROS), eventually leading to cardiomyocyte apoptosis. In addition, OB also inhibited the WNT signaling pathway and downregulated its target genes includinglef1, axin2 and β-catenin. Reactivation of this pathway by the Wnt activator BML-284 and the antioxidant astaxanthin rescued the embryos form the cardiotoxic effects of OB, indicating that oxidative stress, and inhibition of WNT target genes are the mechanistic basis of OB-induced damage in zebrafish. Our study shows that OB exposure causes cardiotoxicity in zebrafish embryos and may be potentially toxic to other aquatic life and even humans.
Collapse
Affiliation(s)
- Yong Huang
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Jinze Ma
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Yunlong Meng
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - You Wei
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Shuling Xie
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Ping Jiang
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Ziqin Wang
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Xiaobei Chen
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Zehui Liu
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Keyuan Zhong
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Zigang Cao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, 343009, Jiangxi, China
| | - Xinjun Liao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, 343009, Jiangxi, China
| | - Juhua Xiao
- Department of Ultrasound, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, 330006, Jiangxi, China
| | - Huiqiang Lu
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, 343009, Jiangxi, China.
| |
Collapse
|
182
|
Mapping Calcium Dynamics in the Heart of Zebrafish Embryos with Ratiometric Genetically Encoded Calcium Indicators. Int J Mol Sci 2020; 21:ijms21186610. [PMID: 32927644 PMCID: PMC7555812 DOI: 10.3390/ijms21186610] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/23/2022] Open
Abstract
Zebrafish embryos have been proposed as a cost-effective vertebrate model to study heart function. Many fluorescent genetically encoded Ca2+ indicators (GECIs) have been developed, but those with ratiometric readout seem more appropriate to image a moving organ such as the heart. Four ratiometric GECIs based on troponin C, TN-XXL, Twitch-1, Twitch-2B, and Twitch-4 were expressed transiently in the heart of zebrafish embryos. Their emission ratio reported the Ca2+ levels in both the atrium and the ventricle. We measured several kinetic parameters of the Ca2+ transients: systolic and diastolic ratio, the amplitude of the systolic Ca2+ rise, the heart rate, as well as the rise and decay times and slopes. The systolic ratio change decreased in cells expressing high biosensor concentration, possibly caused by Ca2+ buffering. The GECIs were able to report the effect of nifedipine and propranolol on the heart, which resulted in changes in heart rate, diastolic and systolic Ca2+ levels, and Ca2+ kinetics. As a result, Twitch-1 and Twitch-4 (Kd 0.25 and 2.8 µM, respectively) seem the most promising GECIs for generating transgenic zebrafish lines, which could be used for modeling heart disorders, for drug screening, and for cardiotoxicity assessment during drug development.
Collapse
|
183
|
Meng Y, Zhong K, Xiao J, Huang Y, Wei Y, Tang L, Chen S, Wu J, Ma J, Cao Z, Liao X, Lu H. Exposure to pyrimethanil induces developmental toxicity and cardiotoxicity in zebrafish. CHEMOSPHERE 2020; 255:126889. [PMID: 32388256 DOI: 10.1016/j.chemosphere.2020.126889] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 05/27/2023]
Abstract
Pyrimethanil is a broad-spectrum fungicide commonly used in the prevention and treatment of Botrytis cinerea. However, little information is available in the literature to show the toxicity of Pyrimethanil to cardiac development. In this study, we used an experimental animal model to explore the developmental and cardiac toxicity of Pyrimethanil in aquatic vertebrates; we exposed zebrafish embryos to Pyrimethanil at concentrations of 2, 4, and 6 mg/L from 5.5 to 72 h post fertilisation. We found that Pyrimethanil caused a decrease in the hatching rate, heart rate, and survival rate of zebrafish embryos. Pyrimethanil exposure also resulted in pericardial and yolk sac edema, spinal deformity, and heart loop failure. Moreover, Pyrimethanil increased reactive oxygen stress levels and heightened the activity of superoxide dismutase and catalase. Alterations were induced in the transcription of apoptosis-related genes (p53, Bax, Bcl2, Casp 9, and Casp6l1) and heart development-related genes (Tbx2b, Gata4, Myh6, Vmhc, Nppa, Bmp2b, Bpm 4, and Bpm 10). Our data showed that the activation of Wnt signalling by BML-284 could partially rescue the malformed phenotype caused by Pyrimethanil. Our results provide new evidence for Pyrimethanil's toxicity and the danger of its residues in the environment and agricultural products.
Collapse
Affiliation(s)
- Yunlong Meng
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Keyuan Zhong
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Juhua Xiao
- Department of Ultrasound, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, 330006, Jiangxi, China
| | - Yong Huang
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - You Wei
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Lin Tang
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Suping Chen
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Juan Wu
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Jinze Ma
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Zigang Cao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, 343009, Jiangxi, China
| | - Xinjun Liao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, 343009, Jiangxi, China
| | - Huiqiang Lu
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, 343009, Jiangxi, China.
| |
Collapse
|
184
|
Cimmino I, Fiory F, Perruolo G, Miele C, Beguinot F, Formisano P, Oriente F. Potential Mechanisms of Bisphenol A (BPA) Contributing to Human Disease. Int J Mol Sci 2020; 21:E5761. [PMID: 32796699 PMCID: PMC7460848 DOI: 10.3390/ijms21165761] [Citation(s) in RCA: 215] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/19/2022] Open
Abstract
Bisphenol A (BPA) is an organic synthetic compound serving as a monomer to produce polycarbonate plastic, widely used in the packaging for food and drinks, medical devices, thermal paper, and dental materials. BPA can contaminate food, beverage, air, and soil. It accumulates in several human tissues and organs and is potentially harmful to human health through different molecular mechanisms. Due to its hormone-like properties, BPA may bind to estrogen receptors, thereby affecting both body weight and tumorigenesis. BPA may also affect metabolism and cancer progression, by interacting with GPR30, and may impair male reproductive function, by binding to androgen receptors. Several transcription factors, including PPARγ, C/EBP, Nrf2, HOX, and HAND2, are involved in BPA action on fat and liver homeostasis, the cardiovascular system, and cancer. Finally, epigenetic changes, such as DNA methylation, histones modification, and changes in microRNAs expression contribute to BPA pathological effects. This review aims to provide an extensive and comprehensive analysis of the most recent evidence about the potential mechanisms by which BPA affects human health.
Collapse
Affiliation(s)
| | | | | | | | | | - Pietro Formisano
- Department of Translational Medicine, Federico II University of Naples and URT “Genomic of Diabetes” of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), 80131 Naples, Italy; (I.C.); (F.F.); (G.P.); (C.M.); (F.B.); (F.O.)
| | | |
Collapse
|
185
|
Modulation and Protection Effects of Antioxidant Compounds against Oxidant Induced Developmental Toxicity in Zebrafish. Antioxidants (Basel) 2020; 9:antiox9080721. [PMID: 32784515 PMCID: PMC7463582 DOI: 10.3390/antiox9080721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022] Open
Abstract
The antioxidant effect of compounds is regularly evaluated by in vitro assays that do not have the capability to predict in vivo protective activity or to determine their underlying mechanisms of action. The aim of this study was to develop an experimental system to evaluate the in vivo protective effects of different antioxidant compounds, based on the zebrafish embryo test. Zebrafish embryos were exposed to tert-butyl hydroperoxide (tBOOH), tetrachlorohydroquinone (TCHQ) and lipopolysaccharides from Escherichia coli (LPS), chemicals that are known inducers of oxidative stress in zebrafish. The developmental toxic effects (lethality or dysmorphogenesis) induced by these chemicals were modulated with n-acetyl l-cysteine and Nω-nitro l-arginine methyl ester hydrochloride, dimethyl maleate and dl-buthionine sulfoximine in order to validate the oxidant mechanism of oxidative stress inducers. The oxidant effects of tBOOH, TCHQ, and LPS were confirmed by the determination of significant differences in the comparison between the concentration–response curves of the oxidative stress inducers and of the modulators of antioxidant status. This concept was also applied to the study of the effects of well-known antioxidants, such as vitamin E, quercetin, and lipoic acid. Our results confirm the zebrafish model as an in vivo useful tool to test the protective effects of antioxidant compounds.
Collapse
|
186
|
Chousidis I, Chatzimitakos T, Leonardos D, Filiou MD, Stalikas CD, Leonardos ID. Cannabinol in the spotlight: Toxicometabolomic study and behavioral analysis of zebrafish embryos exposed to the unknown cannabinoid. CHEMOSPHERE 2020; 252:126417. [PMID: 32200177 DOI: 10.1016/j.chemosphere.2020.126417] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 06/10/2023]
Abstract
Cannabinol (CBN) is a degradation product of the cannabis metabolite Δ9-tetrahydrocannabinol. The CBN concentration in cannabis leaves ranges between 0.1 and 1.6% (w/w of dry weight); it increases as the plant ages and its formation is affected by the storage conditions. As CBN has not been extensively studied so far, the need to examine its impact in vivo is imperative due to the increasing use of cannabis globally. In the study herein, the CBN toxicity, effects on heart physiology, morphological malformations, behavioral changes and alterations in metabolic pathways of zebrafish larvae upon CBN exposure to sublethal concentrations were examined. The LD50 value was estimated at 1.12 mg/l. At the same time, malformations in zebrafish larvae increased significantly in a dose-dependent manner and exposure to CBN concentrations greater than 0.75 mg/l provoked abnormalities like pericardial edema, yolk sac anomalies and tail bending. Concentrations above this threshold resulted in elongated and shorter in width hearts and in separation of ventricle from atrium. The total movement distance and velocity were increased in dark and decreased in light conditions, in a concentration-dependent manner. Our results showed that CBN acts both as a stimulant and a sedative, with larvae to exhibit altered velocity and bradycardia, respectively. The metabolomic analysis revealed alterations mainly to amino acids, which are related to acute toxicity and hint towards systemic metabolic and neuropathophysiological changes. Taken together, our data indicate increased toxic effects as CBN exposure concentration increases, which should be taken into consideration when studying the impact of cannabis on organisms.
Collapse
Affiliation(s)
- Ieremias Chousidis
- Laboratory of Zoology, Biological Applications and Technology Department, University of Ioannina, 45110, Greece
| | - Theodoros Chatzimitakos
- Laboratory of Analytical Chemistry, Department of Chemistry, University of Ioannina, 45110, Greece
| | | | - Michaela D Filiou
- Laboratory of Biochemistry, Biological Applications and Technology Department, University of Ioannina, 45110, Greece
| | - Constantine D Stalikas
- Laboratory of Analytical Chemistry, Department of Chemistry, University of Ioannina, 45110, Greece
| | - Ioannis D Leonardos
- Laboratory of Zoology, Biological Applications and Technology Department, University of Ioannina, 45110, Greece.
| |
Collapse
|
187
|
Mousavi SE, Patil JG. Light-cardiogram, a simple technique for heart rate determination in adult zebrafish, Danio rerio. Comp Biochem Physiol A Mol Integr Physiol 2020; 246:110705. [DOI: 10.1016/j.cbpa.2020.110705] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/26/2020] [Accepted: 04/07/2020] [Indexed: 12/24/2022]
|
188
|
Guzzolino E, Pellegrino M, Ahuja N, Garrity D, D'Aurizio R, Groth M, Baumgart M, Hatcher CJ, Mercatanti A, Evangelista M, Ippolito C, Tognoni E, Fukuda R, Lionetti V, Pellegrini M, Cremisi F, Pitto L. miR-182-5p is an evolutionarily conserved Tbx5 effector that impacts cardiac development and electrical activity in zebrafish. Cell Mol Life Sci 2020; 77:3215-3229. [PMID: 31686119 PMCID: PMC11104936 DOI: 10.1007/s00018-019-03343-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 10/14/2019] [Indexed: 12/20/2022]
Abstract
To dissect the TBX5 regulatory circuit, we focused on microRNAs (miRNAs) that collectively contribute to make TBX5 a pivotal cardiac regulator. We profiled miRNAs in hearts isolated from wild-type, CRE, Tbx5lox/+and Tbx5del/+ mice using a Next Generation Sequencing (NGS) approach. TBX5 deficiency in cardiomyocytes increased the expression of the miR-183 cluster family that is controlled by Kruppel-like factor 4, a transcription factor repressed by TBX5. MiR-182-5p, the most highly expressed miRNA of this family, was functionally analyzed in zebrafish. Transient overexpression of miR-182-5p affected heart morphology, calcium handling and the onset of arrhythmias as detected by ECG tracings. Accordingly, several calcium channel proteins identified as putative miR-182-5p targets were downregulated in miR-182-5p overexpressing hearts. In stable zebrafish transgenic lines, we demonstrated that selective miRNA-182-5p upregulation contributes to arrhythmias. Moreover, cardiac-specific down-regulation of miR-182-5p rescued cardiac defects in a zebrafish model of Holt-Oram syndrome. In conclusion, miR-182-5p exerts an evolutionarily conserved role as a TBX5 effector in the onset of cardiac propensity for arrhythmia, and constitutes a relevant target for mediating the relationship between TBX5, arrhythmia and heart development.
Collapse
Affiliation(s)
- Elena Guzzolino
- Institute of Clinical Physiology, National Research Council, IFC via Moruzzi 1, 56124, Pisa, Italy
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | | | - Neha Ahuja
- Department of Biology, Colorado State University (CSU), Fort Collins, CO, USA
| | - Deborah Garrity
- Department of Biology, Colorado State University (CSU), Fort Collins, CO, USA
| | | | - Marco Groth
- The Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Mario Baumgart
- The Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Cathy J Hatcher
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Alberto Mercatanti
- Institute of Clinical Physiology, National Research Council, IFC via Moruzzi 1, 56124, Pisa, Italy
| | - Monica Evangelista
- Institute of Clinical Physiology, National Research Council, IFC via Moruzzi 1, 56124, Pisa, Italy
| | - Chiara Ippolito
- Department of Clinical and Experimental Medicine, University of Pisa, 56126, Pisa, Italy
| | | | - Ryuichi Fukuda
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Vincenzo Lionetti
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- UOS Anesthesiology, Fondazione Toscana "G.Monasterio", Pisa, Italy
| | | | | | - Letizia Pitto
- Institute of Clinical Physiology, National Research Council, IFC via Moruzzi 1, 56124, Pisa, Italy.
| |
Collapse
|
189
|
Romano N, Ceci M. Are microRNAs responsible for cardiac hypertrophy in fish and mammals? What we can learn in the activation process in a zebrafish ex vivo model. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165896. [PMID: 32681863 DOI: 10.1016/j.bbadis.2020.165896] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/09/2020] [Accepted: 07/11/2020] [Indexed: 02/07/2023]
Abstract
Recent studies have correlated dysregulated miRNA expression with diseased hearts. With the aim of developing an easily manipulated experimental model, phenylephrine (PE) was added to cultured zebrafish hearts to study the expression of miR1 and miR133a by qRT-PCR. Both miRs were downregulated, with greater downregulation leading to higher hypertrophy. The involvement of this miRs was confirmed by the in-vivo inoculation of complementary sequences (AmiR1 and AmiR133a). HSP70 (involved in transporting proteins and in anti-apoptosis processes) was increased in both treatments. Hyperplasia was observed in the epicardium based on WT1 expression (embryonic epicardial cell marker) in both the PE treatment and AmiR133a treatment. The treatment with AmiR1 showed only cardiomyocyte hypertrophy. This ex-vivo model revealed that miR1 and miR133a play a key role in activating early processes leading to myocardium hypertrophy and epicardium hyperplasia and confirmed the expected similarities with hypertrophic disease that occurs in humans.
Collapse
Affiliation(s)
- Nicla Romano
- Dept of Ecology & Biology Sciences, University of Tuscia, Viterbo, Italy.
| | - Marcello Ceci
- Dept of Ecology & Biology Sciences, University of Tuscia, Viterbo, Italy
| |
Collapse
|
190
|
Taufek NM, Harith HH, Abd Rahim MH, Ilham Z, Rowan N, Wan-Mohtar WAAQI. Performance of mycelial biomass and exopolysaccharide from Malaysian Ganoderma lucidum for the fungivore red hybrid Tilapia (Oreochromis sp.) in Zebrafish embryo. AQUACULTURE REPORTS 2020; 17:100322. [DOI: 10.1016/j.aqrep.2020.100322] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
191
|
Machikhin AS, Burlakov AB, Volkov MV, Khokhlov DD. Imaging photoplethysmography and videocapillaroscopy enable noninvasive study of zebrafish cardiovascular system functioning. JOURNAL OF BIOPHOTONICS 2020; 13:e202000061. [PMID: 32306547 DOI: 10.1002/jbio.202000061] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/02/2020] [Accepted: 04/15/2020] [Indexed: 06/11/2023]
Abstract
We report on the noninvasive method for in vivo study of fish's cardiovascular system, that is, the heart and the structure of vessels that carry blood throughout the body. The proposed approach is based on combined photoplethysmographic and videocapillaroscopic microscopic imaging and enables noncontact two-dimensional mapping of blood volume changes. We demonstrate that the obtained data allows precise measurements of heartbeat, blood flow velocity and other important parameters (see Videos S1 and S2). To validate the developed image processing technique, we have carried out multiple experiments on zebrafish-a well-proven informative model organism widely used to understand cardiac development. The proposed approach may be effective for the study of cardiovascular system formation and functioning as well as the impact of various influencing factors on them.
Collapse
Affiliation(s)
- Alexander S Machikhin
- Laboratory of Acoustooptical Spectroscopy, Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
| | - Alexander B Burlakov
- Department of Ichthyology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Mikhail V Volkov
- Department of Applied Optics, University ITMO, Saint Petersburg, Russia
| | - Demid D Khokhlov
- Laboratory of Acoustooptical Spectroscopy, Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
192
|
Chen Z, Yu C, Khan IA, Tang Y, Liu S, Yang M. Toxic effects of different-sized graphene oxide particles on zebrafish embryonic development. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 197:110608. [PMID: 32305822 DOI: 10.1016/j.ecoenv.2020.110608] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 03/28/2020] [Accepted: 04/07/2020] [Indexed: 05/14/2023]
Abstract
Graphene oxide (GO) has broad application potential in many fields, such as biomedicine and energy. Due to the wide-ranging GO applications, its entry into the environment is inevitable along with the potential for ecological and environmental risks. In the present study, we systematically investigated the dose-dependent effects of three different-sized GO particles (50-200 nm, <500 nm, and >500 nm) on zebrafish during the very early developmental stages (4-124 h post-fertilization). The results showed that GOs could accumulate in the eyes, heart, yolk sac, and blood vessels of fish larvae. Consequently, their effects on multiple toxic endpoints were observed, including delayed hatching times, shortened body lengths, alterations in heart rate and blood flow, changes in swimming activity and responses to photoperiod stimulation, and the enhanced activity of total superoxide dismutase, inducible nitric oxide synthase, acetylcholinesterase, caspase-3, and induction of apoptosis-related gene expression. As a result, the occurrence of oxidative stress and the induction of apoptosis are suggested in fish larvae exposed to all three different-sized GO particles. In addition, our results highlight the impacts of waterborne-GO exposure on zebrafish during early development, which were not merely dependent on GO concentration but also on the associated GO sizes. This study hereby provides a basis for the potential ecological and health risks of GO exposure.
Collapse
Affiliation(s)
- Zhong Chen
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China; Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 201306, China; Affiliated Sixth People's Hospital East, Shanghai University of Medicine and Health Sciences, Shanghai, 201306, China.
| | - Cui Yu
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China; School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Imran Ahamed Khan
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China; Shanghai Applied Radiation Institute, Shanghai University, Shanghai, 200444, China
| | - Yi Tang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China; Shanghai Applied Radiation Institute, Shanghai University, Shanghai, 200444, China
| | - Shuai Liu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China; Shanghai Applied Radiation Institute, Shanghai University, Shanghai, 200444, China
| | - Ming Yang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China; Shanghai Applied Radiation Institute, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
193
|
Sun Y, Cao Y, Tong L, Tao F, Wang X, Wu H, Wang M. Exposure to prothioconazole induces developmental toxicity and cardiovascular effects on zebrafish embryo. CHEMOSPHERE 2020; 251:126418. [PMID: 32443233 DOI: 10.1016/j.chemosphere.2020.126418] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 06/11/2023]
Abstract
Prothioconazole is a fungicide that has been widely used in general agriculture and livestock husbandry. This study evaluated the acute toxicity of prothioconazole to zebrafish embryos by assessing their hatching rate and malformation when exposed to different concentrations of prothioconazole. The 96 h-LC50 value of zebrafish embryos was 1.70 mg/L. Upon exposure to 0.85 mg/L, the mortality rate of the embryos significantly increased while their hatching rate decreased significantly. At prothioconazole concentrations higher than 0.43 mg/L, developmental morphologic abnormalities such as heart and yolk-sac edema, spine curvature, tail deformity, shortened body length and decreased eye area were observed. The heart rate of embryos decreased in a dose-dependent fashion during the exposure time. Prothioconazole exposure also resulted in increased rates of cardiac malformation detected by significant increase in the distance between the sinus venosus and bulbus arteriosus and the pericardium area. Moreover, the expression levels of genes related to cardiac development (amhc, vmhc, fli1, hand2, gata4, nkx2.5, tbx5 and atp2a2a) were significantly altered after exposure to prothioconazole. Indeed, this study revealed the adverse effects on the developmental and cardiovascular system of zebrafish embryo caused by prothioconazole. It further elucidated the risk of prothioconazole exposure to vertebrate cardiovascular toxicity. As such, it provides a theoretical foundation for pesticide risk management measures.
Collapse
Affiliation(s)
- Yongqi Sun
- School of Agricultural and Food Science, Zhejiang Agriculture & Forestry University, Hangzhou, China
| | - Yi Cao
- School of Agricultural and Food Science, Zhejiang Agriculture & Forestry University, Hangzhou, China
| | - Lili Tong
- School of Agricultural and Food Science, Zhejiang Agriculture & Forestry University, Hangzhou, China
| | - Fangyi Tao
- School of Agricultural and Food Science, Zhejiang Agriculture & Forestry University, Hangzhou, China
| | - Xiaonan Wang
- School of Agricultural and Food Science, Zhejiang Agriculture & Forestry University, Hangzhou, China
| | - Huiming Wu
- School of Agricultural and Food Science, Zhejiang Agriculture & Forestry University, Hangzhou, China.
| | - Mengcen Wang
- Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Pesticide & Environmental Toxicology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
194
|
Wang M, Shi Y, Yao L, Li Q, Wang Y, Fu D. Potential Molecular Mechanisms and Drugs for Aconitine-Induced Cardiotoxicity in Zebrafish through RNA Sequencing and Bioinformatics Analysis. Med Sci Monit 2020; 26:e924092. [PMID: 32598336 PMCID: PMC7341694 DOI: 10.12659/msm.924092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Accumulating evidence suggests that cardiotoxicity is one of the main manifestations of aconitine (AC) poisoning. However, the molecular mechanism of AC-induced cardiotoxicity remains unclear, there is little direct evidence for therapeutic targets and drugs of AC-induced cardiotoxicity. Material/Methods Zebrafish were exposed to AC to evaluate cardiotoxicity by calculating the heart rates and observing the changes of cardiac and vascular structure. RNA-seq (RNA sequencing) and bioinformatics analysis were used to obtain differentially expressed genes (DEGs). The anti-AC cardiotoxicity compound was identified via connectivity map (CMAP) analysis and molecular docking. Results AC-induced cardiotoxicity in zebrafish predominantly included arrhythmias, extended sinus venous and bulbus arteriosus (SV-BA) distance, and larger pericardial edema aera. A total of 1380 DEGs were identified by RNA-seq and bioinformatics analysis. cyclin-dependent kinase-1 (CDK1) was screened as the hub gene and the most potential therapeutic target due to its significant downregulation in cardiotoxicity based on protein-protein interaction (PPI) and drug-gene interaction (DGIdb) network analysis. Cell cycle signal pathway was the most significant pathways identified in the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Furthermore, the expression of CDK1 was validated in the Gene Expression Omnibus (GEO) database GSE71906, GSE65705, and GSE95140. Finally, heptaminol was identified as a novel anti-AC cardiotoxicity compound via CMAP analysis and molecular docking. Conclusions Totally, hub genes and key pathways identified in this study can aid in the understanding of the molecular changes in AC-induced cardiotoxicity. Meanwhile, we provide a systematic method to explore drug toxicity prevention and treatment.
Collapse
Affiliation(s)
- Mingzhu Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China (mainland)
| | - Yanan Shi
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China (mainland)
| | - Lei Yao
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China (mainland)
| | - Qiang Li
- Children's Hospital of Fudan University, Shanghai, China (mainland)
| | - Youhua Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China (mainland)
| | - Deyu Fu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China (mainland)
| |
Collapse
|
195
|
CFTR deficiency causes cardiac dysplasia during zebrafish embryogenesis and is associated with dilated cardiomyopathy. Mech Dev 2020; 163:103627. [PMID: 32574800 DOI: 10.1016/j.mod.2020.103627] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/04/2020] [Accepted: 06/18/2020] [Indexed: 02/08/2023]
Abstract
Mutations in the CFTR gene cause cystic fibrosis (CF) with myocardial dysfunction. However, it remains unknown whether CF-related heart disease is a secondary effect of pulmonary disease, or an intrinsic primary defect in the heart. Here, we used zebrafish, which lack lung tissue, to investigate the role of CFTR in cardiogenesis. Our findings demonstrated that the loss of CFTR impairs cardiac development from the cardiac progenitor stage, resulting in cardiac looping defects, a dilated atrium, pericardial edema, and a decrease in heart rate. Furthermore, we found that cardiac development was perturbed in wild-type embryos treated with a gating-specific CFTR channel inhibitor, CFTRinh-172, at the blastula stage of development, but not at later stages. Gene expression analysis of blastulas indicated that transcript levels, including mRNAs associated with cardiovascular diseases, were significantly altered in embryos derived from cftr mutants relative to controls. To evaluate the role of CFTR in human heart failure, we performed a genetic association study on individuals with dilated cardiomyopathy and found that the I556V mutation in CFTR, which causes a channel defect, was associated with the disease. Similar to other well-studied channel-defective CFTR mutants, CFTR I556V mRNA failed to restore cardiac dysplasia in mutant embryos. The present study revealed an important role for the CFTR ion channel in regulating cardiac development during early embryogenesis, supporting the hypothesis that CF-related heart disease results from an intrinsic primary defect in the heart.
Collapse
|
196
|
Budine TE, de Sena-Tomás C, Williams MLK, Sepich DS, Targoff KL, Solnica-Krezel L. Gon4l/Udu regulates cardiomyocyte proliferation and maintenance of ventricular chamber identity during zebrafish development. Dev Biol 2020; 462:223-234. [PMID: 32272116 PMCID: PMC10318589 DOI: 10.1016/j.ydbio.2020.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 01/26/2020] [Accepted: 03/02/2020] [Indexed: 01/03/2023]
Abstract
Vertebrate heart development requires spatiotemporal regulation of gene expression to specify cardiomyocytes, increase the cardiomyocyte population through proliferation, and to establish and maintain atrial and ventricular cardiac chamber identities. The evolutionarily conserved chromatin factor Gon4-like (Gon4l), encoded by the zebrafish ugly duckling (udu) locus, has previously been implicated in cell proliferation, cell survival, and specification of mesoderm-derived tissues including blood and somites, but its role in heart formation has not been studied. Here we report two distinct roles of Gon4l/Udu in heart development: regulation of cell proliferation and maintenance of ventricular identity. We show that zygotic loss of udu expression causes a significant reduction in cardiomyocyte number at one day post fertilization that becomes exacerbated during later development. We present evidence that the cardiomyocyte deficiency in udu mutants results from reduced cell proliferation, unlike hematopoietic deficiencies attributed to TP53-dependent apoptosis. We also demonstrate that expression of the G1/S-phase cell cycle regulator, cyclin E2 (ccne2), is reduced in udu mutant hearts, and that the Gon4l protein associates with regulatory regions of the ccne2 gene during early embryogenesis. Furthermore, udu mutant hearts exhibit a decrease in the proportion of ventricular cardiomyocytes compared to atrial cardiomyocytes, concomitant with progressive reduction of nkx2.5 expression. We further demonstrate that udu and nkx2.5 interact to maintain the proportion of ventricular cardiomyocytes during development. However, we find that ectopic expression of nkx2.5 is not sufficient to restore ventricular chamber identity suggesting that Gon4l regulates cardiac chamber patterning via multiple pathways. Together, our findings define a novel role for zygotically-expressed Gon4l in coordinating cardiomyocyte proliferation and chamber identity maintenance during cardiac development.
Collapse
Affiliation(s)
- Terin E Budine
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carmen de Sena-Tomás
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Margot L K Williams
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Diane S Sepich
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kimara L Targoff
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Lila Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
197
|
Linnerz T, Hall CJ. The Diverse Roles of Phagocytes During Bacterial and Fungal Infections and Sterile Inflammation: Lessons From Zebrafish. Front Immunol 2020; 11:1094. [PMID: 32582182 PMCID: PMC7289964 DOI: 10.3389/fimmu.2020.01094] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 05/06/2020] [Indexed: 12/23/2022] Open
Abstract
The immediate and natural reaction to both infectious challenges and sterile insults (wounds, tissue trauma or crystal deposition) is an acute inflammatory response. This inflammatory response is mediated by activation of the innate immune system largely comprising professional phagocytes (neutrophils and macrophages). Zebrafish (danio rerio) larvae possess many advantages as a model organism, including their genetic tractability and highly conserved innate immune system. Exploiting these attributes and the live imaging potential of optically transparent zebrafish larvae has greatly contributed to our understanding of how neutrophils and macrophages orchestrate the initiation and resolution phases of inflammatory responses. Numerous bacterial and fungal infection models have been successfully established using zebrafish as an animal model and studies investigating neutrophil and macrophage behavior to sterile insults have also provided unique insights. In this review we highlight how examining the larval zebrafish response to specific bacterial and fungal pathogens has uncovered cellular and molecular mechanisms behind a variety of phagocyte responses, from those that protect the host to those that are detrimental. We also describe how modeling sterile inflammation in larval zebrafish has provided an opportunity to dissect signaling pathways that control the recruitment, and fate, of phagocytes at inflammatory sites. Finally, we briefly discuss some current limitations, and opportunities to improve, the zebrafish model system for studying phagocyte biology.
Collapse
Affiliation(s)
- Tanja Linnerz
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Christopher J Hall
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
198
|
Benslimane FM, Zakaria ZZ, Shurbaji S, Abdelrasool MKA, Al-Badr MAHI, Al Absi ESK, Yalcin HC. Cardiac function and blood flow hemodynamics assessment of zebrafish (Danio rerio) using high-speed video microscopy. Micron 2020; 136:102876. [PMID: 32512409 DOI: 10.1016/j.micron.2020.102876] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 04/04/2020] [Accepted: 04/14/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND In the last few decades, zebrafish (Danio rerio) were introduced as a model organism to investigate human diseases including cardiovascular and neuronal disorders. In most zebrafish investigations, cardiac function and blood flow hemodynamics need to be assessed to study the effects of the interference on the cardiovascular system. For heart function assessment, most important parameters include heart rate, cardiac output, ejection fraction, fractional area change, and fractional shortening. METHODS A 10 s high-speed video of beating heart and flowing blood within major vessels of zebrafish that are less than 5 days post fertilization (dpf) were recorded via a stereo microscope equipped with a high speed camera. The videos were analyzed using MicroZebraLab and image J software for the assessment of cardiac function. RESULTS Using the technique described here, we were able to simply yet effectively assess cardiac function and blood flow dynamics of normal zebrafish embryos. We believe that the practical method presented here will help cardiac researchers using the zebrafish as a model to examine cardiac function by using tools that could be available in their laboratory.
Collapse
Affiliation(s)
| | - Zain Z Zakaria
- Biomedical Research Center, Qatar University, Doha, Qatar; Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Samar Shurbaji
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | | | | | | |
Collapse
|
199
|
Biomechanical Cues Direct Valvulogenesis. J Cardiovasc Dev Dis 2020; 7:jcdd7020018. [PMID: 32438610 PMCID: PMC7345189 DOI: 10.3390/jcdd7020018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/27/2020] [Accepted: 05/12/2020] [Indexed: 12/30/2022] Open
Abstract
The vertebrate embryonic heart initially forms with two chambers, a ventricle and an atrium, separated by the atrioventricular junction. Localized genetic and biomechanical information guides the development of valves, which function to ensure unidirectional blood flow. If the valve development process goes awry, pathology associated with congenital valve defects can ensue. Congenital valve defects (CVD) are estimated to affect 1–2% of the population and can often require a lifetime of treatment. Despite significant clinical interest, molecular genetic mechanisms that direct valve development remain incompletely elucidated. Cells in the developing valve must contend with a dynamic hemodynamic environment. A growing body of research supports the idea that cells in the valve are highly sensitive to biomechanical forces, which cue changes in gene expression required for normal development or for maintenance of the adult valve. This review will focus on mechanotransductive pathways involved in valve development across model species. We highlight current knowledge regarding how cells sense physical forces associated with blood flow and pressure in the forming heart, and summarize how these changes are transduced into genetic and developmental responses. Lastly, we provide perspectives on how altered biomechanical cues may lead to CVD pathogenesis.
Collapse
|
200
|
Jensen B, Christoffels VM. Reptiles as a Model System to Study Heart Development. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037226. [PMID: 31712265 DOI: 10.1101/cshperspect.a037226] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A chambered heart is common to all vertebrates, but reptiles show unparalleled variation in ventricular septation, ranging from almost absent in tuataras to full in crocodilians. Because mammals and birds evolved independently from reptile lineages, studies on reptile development may yield insight into the evolution and development of the full ventricular septum. Compared with reptiles, mammals and birds have evolved several other adaptations, including compact chamber walls and a specialized conduction system. These adaptations appear to have evolved from precursor structures that can be studied in present-day reptiles. The increase in the number of studies on reptile heart development has been greatly facilitated by sequencing of several genomes and the availability of good staging systems. Here, we place reptiles in their phylogenetic context with a focus on features that are primitive when compared with the homologous features of mammals. Further, an outline of major developmental events is given, and variation between reptile species is discussed.
Collapse
Affiliation(s)
- Bjarke Jensen
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC 1105AZ, Amsterdam, The Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC 1105AZ, Amsterdam, The Netherlands
| |
Collapse
|