51
|
Pott A, Shahid M, Köhler D, Pylatiuk C, Weinmann K, Just S, Rottbauer W. Therapeutic Chemical Screen Identifies Phosphatase Inhibitors to Reconstitute PKB Phosphorylation and Cardiac Contractility in ILK-Deficient Zebrafish. Biomolecules 2018; 8:biom8040153. [PMID: 30463267 PMCID: PMC6315389 DOI: 10.3390/biom8040153] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/24/2018] [Accepted: 10/30/2018] [Indexed: 12/12/2022] Open
Abstract
Patients with inherited dilated cardiomyopathy (DCM) often suffer from severe heart failure based on impaired cardiac contractility leading to increased morbidity and mortality. Integrin-linked kinase (ILK) as a part of the cardiac mechanical stretch sensor was found to be an essential genetic regulator of cardiac contractility. Integrin-linked kinase localizes to z-disks and costameres in vertebrate hearts and regulates the activity of the signaling molecule protein kinase B (PKB/Akt) by controlling its phosphorylation. Despite identification of several potential drug targets in the ILK signaling pathway, pharmacological treatment strategies to restore contractile function in ILK-dependent cardiomyopathies have not been established yet. In recent years, the zebrafish has emerged as a valuable experimental system to model human cardiomyopathies as well as a powerful tool for the straightforward high-throughput in vivo small compound screening of therapeutically active substances. Using the ILK deficient zebrafish heart failure mutant main squeeze (msq), which shows reduced PKB phosphorylation and thereby impaired cardiac contractile force, we identified here, in an automated small compound screen, the protein phosphatase inhibitors calyculin A and okadaic acid significantly restoring myocardial contractile function by reconstituting PKB phosphorylation in msq ILK-deficient zebrafish embryos.
Collapse
Affiliation(s)
- Alexander Pott
- Department of Internal Medicine II, Ulm University, Albert-Einstein-Allee 23, D-89081 Ulm, Germany.
| | - Maryam Shahid
- Department of Internal Medicine II, Ulm University, Albert-Einstein-Allee 23, D-89081 Ulm, Germany.
| | - Doreen Köhler
- Department of Internal Medicine III, University of Heidelberg, D-69120 Heidelberg, Germany.
| | - Christian Pylatiuk
- Institute of Applied Computer Science, Karlsruhe Institute of Technology, D-76344 Eggenstein-Leopoldshafen, Germany.
| | - Karolina Weinmann
- Department of Internal Medicine II, Ulm University, Albert-Einstein-Allee 23, D-89081 Ulm, Germany.
| | - Steffen Just
- Department of Internal Medicine II, Ulm University, Albert-Einstein-Allee 23, D-89081 Ulm, Germany.
| | - Wolfgang Rottbauer
- Department of Internal Medicine II, Ulm University, Albert-Einstein-Allee 23, D-89081 Ulm, Germany.
| |
Collapse
|
52
|
Using Zebrafish for Investigating the Molecular Mechanisms of Drug-Induced Cardiotoxicity. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1642684. [PMID: 30363733 PMCID: PMC6180974 DOI: 10.1155/2018/1642684] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/31/2018] [Accepted: 08/18/2018] [Indexed: 01/09/2023]
Abstract
Over the last decade, the zebrafish (Danio rerio) has emerged as a model organism for cardiovascular research. Zebrafish have several advantages over mammalian models. For instance, the experimental cost of using zebrafish is comparatively low; the embryos are transparent, develop externally, and have high fecundity making them suitable for large-scale genetic screening. More recently, zebrafish embryos have been used for the screening of a variety of toxic agents, particularly for cardiotoxicity testing. Zebrafish has been shown to exhibit physiological responses that are similar to mammals after exposure to medicinal drugs including xenobiotics, hormones, cancer drugs, and also environmental pollutants, including pesticides and heavy metals. In this review, we provided a summary for recent studies that have used zebrafish to investigate the molecular mechanisms of drug-induced cardiotoxicity. More specifically, we focused on the techniques that were exploited by us and others for cardiovascular toxicity assessment and described several microscopic imaging and analysis protocols that are being used for the estimation of a variety of cardiac hemodynamic parameters.
Collapse
|
53
|
Pierucci F, Frati A, Battistini C, Matteini F, Iachini MC, Vestri A, Penna F, Costelli P, Meacci E. Involvement of released sphingosine 1-phosphate/sphingosine 1-phosphate receptor axis in skeletal muscle atrophy. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3598-3614. [PMID: 30279138 DOI: 10.1016/j.bbadis.2018.08.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 07/27/2018] [Accepted: 08/30/2018] [Indexed: 12/18/2022]
Abstract
Skeletal muscle (SkM) atrophy is caused by several and heterogeneous conditions, such as cancer, neuromuscular disorders and aging. In most types of SkM atrophy overall rates of protein synthesis are suppressed, protein degradation is consistently elevated and atrogenes, such as the ubiquitin ligase Atrogin-1/MAFbx, are up-regulated. The molecular regulators of SkM waste are multiple and only in part known. Sphingolipids represent a class of bioactive molecules capable of modulating the destiny of many cell types, including SkM cells. In particular, we and others have shown that sphingosine 1phosphate (S1P), formed by sphingosine kinase (SphK), is able to act as trophic and morphogenic factor in myoblasts. Here, we report the first evidence that the atrophic phenotype observed in both muscle obtained from mice bearing the C26 adenocarcinoma and C2C12 myotubes treated with dexamethasone was characterized by reduced levels of active phospho-SphK1. The importance of SphK1 activity is also confirmed by the specific pharmacological inhibition of SphK1 able to increase Atrogin-1/MAFbx expression and reduce myotube size and myonuclei number. Furthermore, we found that SkM atrophy was accomplished by significant increase of S1P transporter Spns2 and in changes in the pattern of S1P receptor (S1PRs) subtype expression paralleled by increased Atrogin-1/MAFbx expression, suggesting a role for the released S1P and of specific S1PR-mediated signaling pathways in the control of the ubiquitin ligase. Altogether, these findings provide the first evidence that SphK1/released S1P/S1PR axis acts as a molecular regulator of SkM atrophy, thereby representing a new possible target for therapy in many patho-physiological conditions.
Collapse
Affiliation(s)
- Federica Pierucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Alessia Frati
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Chiara Battistini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Francesca Matteini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Maria Chiara Iachini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Ambra Vestri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano (TO), Italy
| | - Paola Costelli
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano (TO), Italy
| | - Elisabetta Meacci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy.
| |
Collapse
|
54
|
Nogee JM, Jay PY. The Heritable Basis of Congenital Heart Disease: Past, Present, and Future. ACTA ACUST UNITED AC 2018; 9:315-7. [PMID: 27531915 DOI: 10.1161/circgenetics.116.001559] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Julie M Nogee
- From the Department of Pediatrics (J.M.N., P.Y.J.) and Department of Genetics (P.Y.J.), Washington University School of Medicine, St. Louis, MO
| | - Patrick Y Jay
- From the Department of Pediatrics (J.M.N., P.Y.J.) and Department of Genetics (P.Y.J.), Washington University School of Medicine, St. Louis, MO.
| |
Collapse
|
55
|
Ultracytochemical visualization of calcium distribution in heart cells and erythrocytes of zebrafish Danio rerio. Micron 2018; 111:19-27. [DOI: 10.1016/j.micron.2018.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 05/23/2018] [Accepted: 05/23/2018] [Indexed: 01/06/2023]
|
56
|
Xu QH, Guan P, Zhang T, Lu C, Li G, Liu JX. Silver nanoparticles impair zebrafish skeletal and cardiac myofibrillogenesis and sarcomere formation. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2018; 200:102-113. [PMID: 29729476 DOI: 10.1016/j.aquatox.2018.04.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 06/08/2023]
Abstract
Metal nanoparticles from industries contaminate the environment and affect the normal development of fish even human health. However, little is known about their biological effects on fish embryogenesis and the potential mechanisms. In this study, zebrafish embryos exposed to/injected with silver nanopaticles (AgNPs) exhibited shorter body, reduced heartbeats, and dysfunctional movements. Less, loose, and unassembled myofibrils were observed in AgNPs-treated embryos, and genes in myofibrillogenesis and sarcomere formation were found to be down-regulated in treated embryos. Down-regulated calcium (Ca2+) signaling and loci-specific DNA methylation in specific muscle genes, such as bves, shroom1, and arpc1a, occurred in AgNPs-treated embryos, which might result in the down-regulated expression of myofibrillogenesis genes and muscle dysfunctions in the treated embryos. Our results for the first time reveal that through down-regulating Ca2+ signaling and myogenic loci-specific DNA methylation in zebrafish embryos, AgNPs might induce defects of myofibril assembly and sarcomere formation via their particles mostly, which may subsequently cause heartbeat reduction and behavior dysfunctions.
Collapse
Affiliation(s)
- Qin-Han Xu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China.
| | - PengPeng Guan
- College of Informatics, Agricultural Bioinformatics Key Laboratory of Hubei Province, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Ting Zhang
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Chang Lu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China.
| | - GuoLiang Li
- College of Informatics, Agricultural Bioinformatics Key Laboratory of Hubei Province, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Jing-Xia Liu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China; Collaborative Innovation Center for Efficient and Health Production of Fisheries in Hunan Province, Hunan, Changde, 415000, China.
| |
Collapse
|
57
|
Zebrafish as a model to study bone maturation: Nanoscale structural and mechanical characterization of age-related changes in the zebrafish vertebral column. J Mech Behav Biomed Mater 2018; 84:54-63. [PMID: 29747057 DOI: 10.1016/j.jmbbm.2018.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/27/2018] [Accepted: 05/03/2018] [Indexed: 11/23/2022]
Abstract
Zebrafish (Danio rerio) is a useful model for understanding biomedical properties of bone and are widely employed in developmental and genetics studies. Here, we have studied the development of zebrafish vertebral bone at the nanoscale from adolescence (6 months), early adulthood (10 months) to mid-life (14 months). Characterization of the bone was conducted using energy-dispersive X-ray spectroscopy (EDX), scanning electron microscopy (SEM) and PeakForce QNM atomic force microscopy (AFM) techniques. SEM and AFM revealed a lamellar structure with mineralized collagen fibrils. There was a significant increase in the wall thickness from 6 to 10 months (76%) and 10 months to 14 months (26%), which is positively correlated with nanomechanical behavior. An increase in the Ca/P ratio was found which was also positively correlated with nanomechanical properties. The change in mechanical properties and Ca/P are similar to those expected in humans when transitioning from adolescence to mid-life. We suggest that zebrafish serve as a suitable model for further studies on age-related changes in bone ultrastructure.
Collapse
|
58
|
Tu HC, Lee GH, Hsiao TH, Kao TT, Wang TY, Tsai JN, Fu TF. One crisis, diverse impacts-Tissue-specificity of folate deficiency-induced circulation defects in zebrafish larvae. PLoS One 2017; 12:e0188585. [PMID: 29176804 PMCID: PMC5703520 DOI: 10.1371/journal.pone.0188585] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/09/2017] [Indexed: 12/17/2022] Open
Abstract
Folate (vitamin B9) is an essential nutrient required for cell survival, proliferation, differentiation and therefore embryogenesis. Folate deficiency has been associated with many diseases, including congenital heart diseases and megaloblastic anemia, yet the mechanisms underlying these remains elusive. Here, we examine the impact of folate deficiency on the development of the circulation system using a zebrafish transgenic line which displays inducible folate deficiency. Impaired hematopoiesis includes decreased hemoglobin levels, decreased erythrocyte number, increased erythrocyte size and aberrant c-myb expression pattern were observed in folate deficient embryos. Cardiac defects, including smaller chamber size, aberrant cardiac function and cmlc2 expression pattern, were also apparent in folate deficient embryos. Characterization of intracellular folate content in folate deficiency revealed a differential fluctuation among the different folate derivatives that carry a single carbon group at different oxidation levels. Rescue attempts by folic acid and nucleotides resulted in differential responses among affected tissues, suggesting that different pathomechanisms are involved in folate deficiency-induced anomalies in a tissue-specific manner. The results of the current study provide an explanation for the inconsistent outcome observed clinically in patients suffering from folate deficiency and/or receiving folate supplementation. This study also supports the use of this model for further research on the defective cardiogenesis and hematopoiesis caused by folate deficiency.
Collapse
Affiliation(s)
- Hung-Chi Tu
- The Institute of Basic Medical Sciences, National Cheng Kung University, College of Medicine, Tainan, Taiwan
| | - Gang-Hui Lee
- The Institute of Basic Medical Sciences, National Cheng Kung University, College of Medicine, Tainan, Taiwan
| | - Tsun-Hsien Hsiao
- The Institute of Basic Medical Sciences, National Cheng Kung University, College of Medicine, Tainan, Taiwan
| | - Tseng-Ting Kao
- The Institute of Basic Medical Sciences, National Cheng Kung University, College of Medicine, Tainan, Taiwan
| | - Tzu-Ya Wang
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, College of Medicine, Tainan, Taiwan
| | - Jen-Ning Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Department of Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Tzu-Fun Fu
- The Institute of Basic Medical Sciences, National Cheng Kung University, College of Medicine, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, College of Medicine, Tainan, Taiwan
- * E-mail:
| |
Collapse
|
59
|
Yue C, Ji C, Zhang H, Zhang LW, Tong J, Jiang Y, Chen T. Protective effects of folic acid on PM2.5-induced cardiac developmental toxicity in zebrafish embryos by targeting AhR and Wnt/β-catenin signal pathways. ENVIRONMENTAL TOXICOLOGY 2017; 32:2316-2322. [PMID: 28722335 DOI: 10.1002/tox.22448] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 06/21/2017] [Accepted: 07/02/2017] [Indexed: 05/06/2023]
Abstract
Our previous observations indicated that extractable organic matter (EOM) from PM2.5 induced malformations in the heart of zebrafish embryos by activating AhR and inhibiting canonical Wnt/β-catenin signal pathway. As a nutritional factor, folic acid (FA) is reported to prevent cardiac defects during embryo development. Hence, we hypothesize that FA may prevent PM2.5-induced heart defects by interfering with AhR and Wnt/β-catenin signaling pathways. Our results showed that FA supplementation alleviated the EOM-induced heart defects in zebrafish embryos, and both AhR inhibitor CH223191 and Wnt activator CHIR99021 enhanced the protective efficiency of FA. Furthermore, FA supplementation attenuated the EOM-induced upregulation of AhR and its target genes including Cyp1a1, Cyp1b1, Ahrra, and Ahrrb. EROD assay confirmed that the EOM agonized Cyp1a1 activity was diminished by FA. The EOM-induced downregulation of β-catenin and its target genes including Nkx2.5, Axin2, Sox9b, and Cox2b were recovered or even overexpressed in embryos exposed to EOM plus FA. In conclusion, our study suggested that FA supplementation protected against PM2.5 cardiac development toxicity by targeting AhR and Wnt/β-catenin signal pathways.
Collapse
Affiliation(s)
- Cong Yue
- Department of Toxicology, School of Public Health, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Cheng Ji
- Department of Genetics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Hang Zhang
- Department of Toxicology, School of Public Health, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Leshuai W Zhang
- School for Radiological and interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Jian Tong
- Department of Toxicology, School of Public Health, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Yan Jiang
- Department of Physiology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Tao Chen
- Department of Toxicology, School of Public Health, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| |
Collapse
|
60
|
Harrington JK, Sorabella R, Tercek A, Isler JR, Targoff KL. Nkx2.5 is essential to establish normal heart rate variability in the zebrafish embryo. Am J Physiol Regul Integr Comp Physiol 2017; 313:R265-R271. [PMID: 28615160 DOI: 10.1152/ajpregu.00223.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 12/11/2022]
Abstract
Heart rate variability (HRV) has become an important clinical marker of cardiovascular health and a research measure for the study of the cardiac conduction system and its autonomic controls. While the zebrafish (Danio rerio) is an ideal vertebrate model for understanding heart development, HRV has only recently been investigated in this system. We have previously demonstrated that nkx2.5 and nkx2.7, two homologues of Nkx2-5 expressed in zebrafish cardiomyocytes, play vital roles in maintaining cardiac chamber-specific characteristics. Given observed defects in ventricular and atrial chamber identities in nkx2.5-/- embryos coupled with conduction system abnormalities in murine models of Nkx2.5 insufficiency, we postulated that reduced HRV would serve as a marker of poor cardiac health in nkx2.5 mutants and in other zebrafish models of human congenital heart disease. Using live video image acquisition, we derived beat-to-beat intervals to compare HRV in wild-type and nkx2.5-/- embryos. Our data illustrate that the nkx2.5 loss-of-function model exhibits increased heart rate and decreased HRV when compared with wild type during embryogenesis. These findings validate HRV analysis as a useful quantitative tool for assessment of cardiac health in zebrafish and underscore the importance of nkx2.5 in maintaining normal heart rate and HRV during early conduction system development.
Collapse
Affiliation(s)
- Jamie K Harrington
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Robert Sorabella
- Division of Cardiothoracic Surgery, Department of Surgery, College of Physicians and Surgeons, Columbia University, New York, New York; and
| | - Abigail Tercek
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Joseph R Isler
- Division of Neonatology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Kimara L Targoff
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York;
| |
Collapse
|
61
|
Cyp26 Enzymes Facilitate Second Heart Field Progenitor Addition and Maintenance of Ventricular Integrity. PLoS Biol 2016; 14:e2000504. [PMID: 27893754 PMCID: PMC5125711 DOI: 10.1371/journal.pbio.2000504] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 10/28/2016] [Indexed: 11/19/2022] Open
Abstract
Although retinoic acid (RA) teratogenicity has been investigated for decades, the mechanisms underlying RA-induced outflow tract (OFT) malformations are not understood. Here, we show zebrafish embryos deficient for Cyp26a1 and Cyp26c1 enzymes, which promote RA degradation, have OFT defects resulting from two mechanisms: first, a failure of second heart field (SHF) progenitors to join the OFT, instead contributing to the pharyngeal arch arteries (PAAs), and second, a loss of first heart field (FHF) ventricular cardiomyocytes due to disrupted cell polarity and extrusion from the heart tube. Molecularly, excess RA signaling negatively regulates fibroblast growth factor 8a (fgf8a) expression and positively regulates matrix metalloproteinase 9 (mmp9) expression. Although restoring Fibroblast growth factor (FGF) signaling can partially rescue SHF addition in Cyp26 deficient embryos, attenuating matrix metalloproteinase (MMP) function can rescue both ventricular SHF addition and FHF integrity. These novel findings indicate a primary effect of RA-induced OFT defects is disruption of the extracellular environment, which compromises both SHF recruitment and FHF ventricular integrity. Retinoic acid (RA) is the most active metabolic product of vitamin A. The embryonic heart is particularly sensitive to inappropriate RA levels, with cardiac outflow tract (OFT) defects among the most common RA-induced malformations. However, the mechanisms underlying these RA-induced defects are not understood. Cyp26 enzymes facilitate degradation of RA and thus are required to limit RA levels in early development. Here, we present evidence that loss of Cyp26 enzymes induces cardiac OFT defects through two mechanisms. First, we find that Cyp26-deficient zebrafish embryos fail to add later-differentiating ventricular cardiac progenitors to the OFT, with some of these progenitors instead contributing to the nearby arch arteries. Second, Cyp26-deficient embryos cannot maintain the integrity of the nascent heart tube, with ventricular cells within the heart tube losing their polarity and being extruded. Our data indicate that excess expression of matrix metalloproteinase 9, an enzyme that degrades the extracellular matrix, underlies both the cardiac progenitor addition and heart tube integrity defects seen in Cyp26-deficient embryos. Our findings highlight perturbation of the extracellular matrix as a major cause of RA-induced cardiac OFT defects that specifically disrupt ventricular development at later stages than previously appreciated.
Collapse
|
62
|
Cardiomyocyte proliferation in zebrafish and mammals: lessons for human disease. Cell Mol Life Sci 2016; 74:1367-1378. [PMID: 27812722 PMCID: PMC5357290 DOI: 10.1007/s00018-016-2404-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 10/14/2016] [Accepted: 10/28/2016] [Indexed: 01/08/2023]
Abstract
Cardiomyocytes proliferate profusely during early development and for a brief period after birth in mammals. Within a month after birth, this proliferative capability is dramatically reduced in mammals unlike lower vertebrates where it persists into adult life. The zebrafish, for example, retains the ability to regenerate the apex of the heart following resection by a mechanism predominantly driven by cardiomyocyte proliferation. Differences in proliferative capacity of cardiomyocytes in adulthood between mammals and lower vertebrates are closely liked to ontogenetic or phylogenetic factors. Elucidation of these factors has the potential to provide enormous benefits if they lead to the development of therapeutic strategies that facilitate cardiomyocyte proliferation. In this review, we highlight the differences between Mammalian and Zebrafish cardiomyocytes, which could explain at least in part the different proliferative capacities in these two species. We discuss the advantages of the zebrafish as a model of cardiomyocyte proliferation, particularly at the embryonic stage. We also identify a number of key molecular pathways with potential to reveal key steps in switching cardiomyocytes from a quiescent to a proliferative phenotype.
Collapse
|
63
|
Vargas R, Vásquez IC. Cardiac and somatic parameters in zebrafish: tools for the evaluation of cardiovascular function. FISH PHYSIOLOGY AND BIOCHEMISTRY 2016; 42:569-577. [PMID: 26553553 DOI: 10.1007/s10695-015-0160-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 11/02/2015] [Indexed: 06/05/2023]
Abstract
Cardiovascular diseases are a worldwide public health problem. To date, extensive research has been conducted to elucidate the pathophysiological mechanisms that trigger cardiovascular diseases and to evaluate therapeutic options. Animal models are widely used to achieve these goals, and zebrafish have emerged as a low-cost model that produces rapid results. Currently, a large body of research is devoted to the cardiovascular development and diverse cardiovascular disorders of zebrafish embryos and larvae. However, less research has been conducted on adult zebrafish specimens. In this study, we evaluated a method to obtain and to evaluate morphometric parameters (of both the entire animal and the heart) of adult zebrafish. We used these data to calculate additional parameters, such as body mass index, condition factor and cardiac somatic index. This method and its results can be used as reference for future studies that aim to evaluate the pathophysiological aspects of the zebrafish cardiovascular system.
Collapse
Affiliation(s)
- Rafael Vargas
- Departamento de Ciencias Fisiológicas, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá, Colombia.
| | - Isabel Cristina Vásquez
- Departamento de Ciencias Fisiológicas, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
64
|
Cardiac sodium channel regulator MOG1 regulates cardiac morphogenesis and rhythm. Sci Rep 2016; 6:21538. [PMID: 26903377 PMCID: PMC4763225 DOI: 10.1038/srep21538] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/26/2016] [Indexed: 01/09/2023] Open
Abstract
MOG1 was initially identified as a protein that interacts with the small GTPase Ran involved in transport of macromolecules into and out of the nucleus. In addition, we have established that MOG1 interacts with the cardiac sodium channel Nav1.5 and regulates cell surface trafficking of Nav1.5. Here we used zebrafish as a model system to study the in vivo physiological role of MOG1. Knockdown of mog1 expression in zebrafish embryos significantly decreased the heart rate (HR). Consistently, the HR increases in embryos with over-expression of human MOG1. Compared with wild type MOG1 or control EGFP, mutant MOG1 with mutation E83D associated with Brugada syndrome significantly decreases the HR. Interestingly, knockdown of mog1 resulted in abnormal cardiac looping during embryogenesis. Mechanistically, knockdown of mog1 decreases expression of hcn4 involved in the regulation of the HR, and reduces expression of nkx2.5, gata4 and hand2 involved in cardiac morphogenesis. These data for the first time revealed a novel role that MOG1, a nucleocytoplasmic transport protein, plays in cardiac physiology and development.
Collapse
|
65
|
Kwon HJ. Vitamin D receptor signaling is required for heart development in zebrafish embryo. Biochem Biophys Res Commun 2016; 470:575-578. [PMID: 26797277 DOI: 10.1016/j.bbrc.2016.01.103] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 01/16/2016] [Indexed: 11/29/2022]
Abstract
Vitamin D has been found to be associated with cardiovascular diseases. However, the role of vitamin D in heart development during embryonic period is largely unknown. Vitamin D induces its genomic effects through its nuclear receptor, the vitamin D receptor (VDR). The present study investigated the role of VDR on heart development by antisense-mediated knockdown approaches in zebrafish model system. In zebrafish embryos, two distinct VDR genes (vdra and vdrb) have been identified. Knockdown of vdra has little effect on heart development, whereas disrupting vdrb gene causes various cardiac phenotypes, characterized by pericardial edema, slower heart rate and laterality defects. Depletion of both vdra and vdrb (vdra/b) produce additive, but not synergistic effects. To determine whether atrioventricular (AV) cardiomyocytes are properly organized in these embryos, the expression of bmp4, which marks the developing AV boundary at 48 h post-fertilization, was examined. Notably, vdra/b-deficient embryos display ectopic expression of bmp4 towards the ventricle or throughout atrial and ventricular chambers. Taken together, these results suggest that VDR signaling plays an essential role in heart development.
Collapse
Affiliation(s)
- Hye-Joo Kwon
- Biology Department, Texas A&M University, College Station, TX77843-3258, United States; Biology Department, Princess Nourah University, Riyadh 11671, Saudi Arabia.
| |
Collapse
|
66
|
Stoppel WL, Kaplan DL, Black LD. Electrical and mechanical stimulation of cardiac cells and tissue constructs. Adv Drug Deliv Rev 2016; 96:135-55. [PMID: 26232525 DOI: 10.1016/j.addr.2015.07.009] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/16/2015] [Accepted: 07/25/2015] [Indexed: 12/19/2022]
Abstract
The field of cardiac tissue engineering has made significant strides over the last few decades, highlighted by the development of human cell derived constructs that have shown increasing functional maturity over time, particularly using bioreactor systems to stimulate the constructs. However, the functionality of these tissues is still unable to match that of native cardiac tissue and many of the stem-cell derived cardiomyocytes display an immature, fetal like phenotype. In this review, we seek to elucidate the biological underpinnings of both mechanical and electrical signaling, as identified via studies related to cardiac development and those related to an evaluation of cardiac disease progression. Next, we review the different types of bioreactors developed to individually deliver electrical and mechanical stimulation to cardiomyocytes in vitro in both two and three-dimensional tissue platforms. Reactors and culture conditions that promote functional cardiomyogenesis in vitro are also highlighted. We then cover the more recent work in the development of bioreactors that combine electrical and mechanical stimulation in order to mimic the complex signaling environment present in vivo. We conclude by offering our impressions on the important next steps for physiologically relevant mechanical and electrical stimulation of cardiac cells and engineered tissue in vitro.
Collapse
|
67
|
Braunbeck T, Kais B, Lammer E, Otte J, Schneider K, Stengel D, Strecker R. The fish embryo test (FET): origin, applications, and future. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2015; 22:16247-61. [PMID: 25395325 DOI: 10.1007/s11356-014-3814-7] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 11/03/2014] [Indexed: 05/06/2023]
Abstract
Originally designed as an alternative for the acute fish toxicity test according to, e.g., OECD TG 203, the fish embryo test (FET) with the zebrafish (Danio rerio) has been optimized, standardized, and validated during an OECD validation study and adopted as OECD TG 236 as a test to assess toxicity of embryonic forms of fish. Given its excellent correlation with the acute fish toxicity test and the fact that non-feeding developmental stages of fish are not categorized as protected stages according to the new European Directive 2010/63/EU on the protection of animals used for scientific purposes, the FET is ready for use not only for range-finding but also as a true alternative for the acute fish toxicity test, as required for a multitude of national and international regulations. If-for ethical reasons-not accepted as a full alternative, the FET represents at least a refinement in the sense of the 3Rs principle. Objections to the use of the FET have mainly been based on the putative lack of biotransformation capacity and the assumption that highly lipophilic and/or high molecular weight substances might not have access to the embryo due to the protective role of the chorion. With respect to bioactivation, the only substance identified so far as not being activated in the zebrafish embryo is allyl alcohol; all other biotransformation processes that have been studied in more detail so far were found to be present, albeit, in some cases, at lower levels than in adult fish. With respect to larger molecules, the extension of the test duration to 96 h (i.e., beyond hatch) has-at least for the substances tested so far-compensated for the reduced access to the embryo; however, more research is necessary to fully explore the applicability of the FET to substances with a molecular weight >3 kDa as well as substances with a neurotoxic mode of action. An extension of the endpoints to also cover sublethal endpoints makes the FET a powerful tool for the detection of teratogenicity, dioxin-like activity, genotoxicity and mutagenicity, neurotoxicity, as well as various forms of endocrine disruption.
Collapse
Affiliation(s)
- Thomas Braunbeck
- Aquatic Ecology and Toxicology Group, Center for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany.
| | - Britta Kais
- Aquatic Ecology and Toxicology Group, Center for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany
| | - Eva Lammer
- Aquatic Ecology and Toxicology Group, Center for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany
| | - Jens Otte
- Aquatic Ecology and Toxicology Group, Center for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany
| | - Katharina Schneider
- Aquatic Ecology and Toxicology Group, Center for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany
| | - Daniel Stengel
- Aquatic Ecology and Toxicology Group, Center for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany
| | - Ruben Strecker
- Aquatic Ecology and Toxicology Group, Center for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany
| |
Collapse
|
68
|
Matrone G, Wilson KS, Mullins JJ, Tucker CS, Denvir MA. Temporal cohesion of the structural, functional and molecular characteristics of the developing zebrafish heart. Differentiation 2015; 89:117-27. [PMID: 26095446 DOI: 10.1016/j.diff.2015.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 04/06/2015] [Accepted: 05/10/2015] [Indexed: 11/25/2022]
Abstract
Heart formation is a complex, dynamic and highly coordinated process of molecular, morphogenetic and functional factors with each interacting and contributing to formation of the mature organ. Cardiac abnormalities in early life can be lethal in mammals but not in the zebrafish embryo which has been widely used to study the developing heart. While early cardiac development in the zebrafish has been well characterized, functional changes during development and how these relate to architectural, cellular and molecular aspects of development have not been well described previously. To address this we have carefully characterised cardiac structure, function, cardiomyocyte proliferation and cardiac-specific gene expression between 48 and 120 hpf in the zebrafish. We show that the zebrafish heart increases in volume and changes shape significantly between 48 and 72 hpf accompanied by a 40% increase in cardiomyocyte number. Between 96 and 120 hpf, while external heart expansion slows, there is rapid formation of a mature and extensive trabecular network within the ventricle chamber. While ejection fraction does not change during the course of development other determinants of contractile function increase significantly particularly between 72 and 96 hpf leading to an increase in cardinal vein blood flow. This study has revealed a number of novel aspects of cardiac developmental dynamics with striking temporal orchestration of structure and function within the first few days of development. These changes are associated with changes in expression of developmental and maturational genes. This study provides important insights into the complex temporal relationship between structure and function of the developing zebrafish heart.
Collapse
Affiliation(s)
- Gianfranco Matrone
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom.
| | - Kathryn S Wilson
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - John J Mullins
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Carl S Tucker
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Martin A Denvir
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| |
Collapse
|
69
|
Kessler EL, van Veen TAB. A fishing trip to cure arrhythmogenic cardiomyopathy? ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:90. [PMID: 26015932 DOI: 10.3978/j.issn.2305-5839.2015.01.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 01/16/2015] [Indexed: 11/14/2022]
Abstract
The paper entitled "Identification of a New Modulator of the Intercalated Disc in a Zebrafish Model of Arrhythmogenic Cardiomyopathy", as published in 2014 in Science Translational Medicine, examined the effects of the newly discovered drug SB216763 (SB21) on arrhythmogenic cardiomyopathy (ACM). In this paper, the authors focused on mechanisms underlying ACM and the accompanying molecular and cellular alterations. Most importantly they showed that SB21 was able to rescue and partly reverse the ACM phenotype in three different experimental models: (I) a zebrafish model of Naxos disease induced by the overexpression of the 2057del2 mutation in plakoglobin (PKG); (II) neonatal rat cardiomyocytes overexpressing the same mutation in PKG; (III) cardiomyocytes derived from induced pluripotent stem cells expressing two different forms of mutations in plakophilin-2. This editorial will focus on the potency and possible restrictions concerning SB21 treatment as a potential intervention for ACM and the usefulness of the applied zebrafish models in general.
Collapse
Affiliation(s)
- Elise L Kessler
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht Utrecht, The Netherlands
| | - Toon A B van Veen
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht Utrecht, The Netherlands
| |
Collapse
|
70
|
Hassinen M, Haverinen J, Hardy ME, Shiels HA, Vornanen M. Inward rectifier potassium current (I K1) and Kir2 composition of the zebrafish (Danio rerio) heart. Pflugers Arch 2015; 467:2437-46. [PMID: 25991088 DOI: 10.1007/s00424-015-1710-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/07/2015] [Accepted: 05/07/2015] [Indexed: 11/24/2022]
Abstract
Electrophysiological properties and molecular background of the zebrafish (Danio rerio) cardiac inward rectifier current (IK1) were examined. Ventricular myocytes of zebrafish have a robust (-6.7 ± 1.2 pA pF(-1) at -120 mV) strongly rectifying and Ba(2+)-sensitive (IC50 = 3.8 μM) IK1. Transcripts of six Kir2 channels (drKir2.1a, drKir2.1b, drKir2.2a, drKir2.2b, drKir2.3, and drKir2.4) were expressed in the zebrafish heart. drKir2.4 and drKir2.2a were the dominant isoforms in both the ventricle (92.9 ± 1.5 and 6.3 ± 1.5%) and the atrium (28.9 ± 2.9 and 64.7 ± 3.0%). The remaining four channels comprised together less than 1 and 7 % of the total transcripts in ventricle and atrium, respectively. The four main gene products (drKir2.1a, drKir2.2a, drKir2.2b, drKir2.4) were cloned, sequenced, and expressed in HEK cells for electrophysiological characterization. drKir2.1a was the most weakly rectifying (passed more outward current) and drKir2.2b the most strongly rectifying (passed less outward current) channel, whilst drKir2.2a and drKir2.4 were intermediate between the two. In regard to sensitivity to Ba(2+) block, drKir2.4 was the most sensitive (IC50 = 1.8 μM) and drKir2.1a the least sensitive channel (IC50 = 132 μM). These findings indicate that the Kir2 isoform composition of the zebrafish heart markedly differs from that of mammalian hearts. Furthermore orthologous Kir2 channels (Kir2.1 and Kir2.4) of zebrafish and mammals show striking differences in Ba(2+)-sensitivity. Structural and functional differences needs to be taken into account when zebrafish is used as a model for human cardiac electrophysiology, cardiac diseases, and in screening cardioactive substances.
Collapse
Affiliation(s)
- Minna Hassinen
- Department of Biology, University of Eastern Finland, P.O. Box 111, 80101, Joensuu, Finland.
| | - Jaakko Haverinen
- Department of Biology, University of Eastern Finland, P.O. Box 111, 80101, Joensuu, Finland
| | - Matt E Hardy
- Faculty of Life Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Holly A Shiels
- Faculty of Life Sciences, University of Manchester, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Matti Vornanen
- Department of Biology, University of Eastern Finland, P.O. Box 111, 80101, Joensuu, Finland
| |
Collapse
|
71
|
Yang J, Shih YH, Xu X. Understanding cardiac sarcomere assembly with zebrafish genetics. Anat Rec (Hoboken) 2015; 297:1681-93. [PMID: 25125181 DOI: 10.1002/ar.22975] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 05/12/2014] [Accepted: 05/13/2014] [Indexed: 01/06/2023]
Abstract
Mutations in sarcomere genes have been found in many inheritable human diseases, including hypertrophic cardiomyopathy. Elucidating the molecular mechanisms of sarcomere assembly shall facilitate understanding of the pathogenesis of sarcomere-based cardiac disease. Recently, biochemical and genomic studies have identified many new genes encoding proteins that localize to the sarcomere. However, their precise functions in sarcomere assembly and sarcomere-based cardiac disease are unknown. Here, we review zebrafish as an emerging vertebrate model for these studies. We summarize the techniques offered by this animal model to manipulate genes of interest, annotate gene expression, and describe the resulting phenotypes. We survey the sarcomere genes that have been investigated in zebrafish and discuss the potential of applying this in vivo model for larger-scale genetic studies.
Collapse
Affiliation(s)
- Jingchun Yang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota; Division of Cardiovascular Diseases, Mayo Clinic College of Medicine, Rochester, Minnesota
| | | | | |
Collapse
|
72
|
Block the function of nonmuscle myosin II by blebbistatin induces zebrafish embryo cardia bifida. In Vitro Cell Dev Biol Anim 2014; 51:211-7. [PMID: 25403653 DOI: 10.1007/s11626-014-9836-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 10/16/2014] [Indexed: 10/24/2022]
Abstract
Nonmuscle myosin II (NM II) is the name given to the multi-subunit protein product of three genes encoding different nonmuscle myosin heavy chains including NM II-A, NM II-B, and NM II-C. Blebbistatin is a small molecule that has been shown to be a relatively specific inhibitor of NM II. Blocking the function of NM II by blebbistatin induces zebrafish embryo cardia bifida at a dose-dependent manner. In situ hybridization analysis with ventricular marker ventricular myosin heavy chain (vmhc) and atrial marker atrial myosin heavy chain (amhc) showed each of the heart contained both distinct atria and ventricle. However, the cardia bifida embryos had highly variable distance between two separate ventricles. We also provided evidence that time window from 12 to 20 h post fertilization (hpf) is necessary and sufficient for cardia bifida formation caused by blebbistatin treatment. Expression of spinster homolog 2 (spns2) was decreased in blebbistatin-treated embryos, suggesting the cardia bifida phenotype caused by NM II inhibition was relevant to precardiac mesoderm migration defects. Through in situ hybridization analysis, we showed that foxa1 was expressed in endoderm of blebbistatin-treated embryos at 24-hpf stage, suggesting the endoderm formation is normal in cardia bifida embryos caused by blebbistatin treatment. In addition, we demonstrated that blebbistatin treatment resulted in morphology alteration of zebrafish cardiomyocytes in vivo and neonatal mouse cardiomyocytes in vitro.
Collapse
|
73
|
Hou JH, Kralj JM, Douglass AD, Engert F, Cohen AE. Simultaneous mapping of membrane voltage and calcium in zebrafish heart in vivo reveals chamber-specific developmental transitions in ionic currents. Front Physiol 2014; 5:344. [PMID: 25309445 PMCID: PMC4161048 DOI: 10.3389/fphys.2014.00344] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 08/22/2014] [Indexed: 01/31/2023] Open
Abstract
The cardiac action potential (AP) and the consequent cytosolic Ca2+ transient are key indicators of cardiac function. Natural developmental processes, as well as many drugs and pathologies change the waveform, propagation, or variability (between cells or over time) of these parameters. Here we apply a genetically encoded dual-function calcium and voltage reporter (CaViar) to study the development of the zebrafish heart in vivo between 1.5 and 4 days post fertilization (dpf). We developed a high-sensitivity spinning disk confocal microscope and associated software for simultaneous three-dimensional optical mapping of voltage and calcium. We produced a transgenic zebrafish line expressing CaViar under control of the heart-specific cmlc2 promoter, and applied ion channel blockers at a series of developmental stages to map the maturation of the action potential in vivo. Early in development, the AP initiated via a calcium current through L-type calcium channels. Between 90 and 102 h post fertilization (hpf), the ventricular AP switched to a sodium-driven upswing, while the atrial AP remained calcium driven. In the adult zebrafish heart, a sodium current drives the AP in both the atrium and ventricle. Simultaneous voltage and calcium imaging with genetically encoded reporters provides a new approach for monitoring cardiac development, and the effects of drugs on cardiac function.
Collapse
Affiliation(s)
- Jennifer H Hou
- Department of Physics, Harvard University Cambridge, MA, USA
| | - Joel M Kralj
- Department of Chemistry and Chemical Biology, Harvard University Cambridge, MA, USA
| | - Adam D Douglass
- Department of Molecular and Cellular Biology, Harvard University Cambridge, MA, USA
| | - Florian Engert
- Department of Molecular and Cellular Biology, Harvard University Cambridge, MA, USA
| | - Adam E Cohen
- Department of Physics, Harvard University Cambridge, MA, USA ; Department of Chemistry and Chemical Biology, Harvard University Cambridge, MA, USA ; Howard Hughes Medical Institute Chevy Chase, MD, USA
| |
Collapse
|
74
|
Abstract
A notable advantage of zebrafish as a model organism is the ease of gene knockdown using morpholino antisense oligonucleotide (MO). However, zebrafish morphants injected with MO for a target protein often show heterogeneous phenotypes, despite controlling the injection volume of the MO solution in all embryos. We developed a method for estimating the quantity of MO injected into each living morphant, based on the co-injection of a control MO labeled with the fluorophore lissamine. By applying this method for knockdown of cardiac troponin T (tnnt2a) in zebrafish, we could efficiently select the partial tnnt2a-depleted zebrafish with a decreased heart rate and impairment of cardiac contraction. To investigate cardiac impairment of the tnnt2a morphant, we performed fluorescent cardiac imaging using Bodipy-ceramide. Cardiac image analysis showed moderate reduction of tnnt2a impaired diastolic distensibility and decreased contraction and relaxation velocities. To the best of our knowledge, this is the first report to analyze the role of tnnt2a in cardiac function in tnnt2a-depleted living animals. Our combinatorial approach can be applied for analyzing the molecular function of any protein associated with human cardiac diseases.
Collapse
|
75
|
Dickover M, Hegarty JM, Ly K, Lopez D, Yang H, Zhang R, Tedeschi N, Hsiai TK, Chi NC. The atypical Rho GTPase, RhoU, regulates cell-adhesion molecules during cardiac morphogenesis. Dev Biol 2014; 389:182-91. [PMID: 24607366 DOI: 10.1016/j.ydbio.2014.02.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 01/28/2014] [Accepted: 02/18/2014] [Indexed: 11/29/2022]
Abstract
The vertebrate heart undergoes early complex morphologic events in order to develop key cardiac structures that regulate its overall function (Fahed et al., 2013). Although many genetic factors that participate in patterning the heart have been elucidated (Tu and Chi, 2012), the cellular events that drive cardiac morphogenesis have been less clear. From a chemical genetic screen to identify cellular pathways that control cardiac morphogenesis in zebrafish, we observed that inhibition of the Rho signaling pathways resulted in failure to form the atrioventricular canal and loop the linear heart tube. To identify specific Rho proteins that may regulate this process, we analyzed cardiac expression profiling data and discovered that RhoU was expressed at the atrioventricular canal during the time when it forms. Loss of RhoU function recapitulated the atrioventricular canal and cardiac looping defects observed in the ROCK inhibitor treated zebrafish. Similar to its family member RhoV/Chp (Tay et al., 2010), we discovered that RhoU regulates the cell junctions between cardiomyocytes through the Arhgef7b/Pak kinase pathway in order to guide atrioventricular canal development and cardiac looping. Inhibition of this pathway resulted in similar underlying cardiac defects and conversely, overexpression of a PAK kinase was able to rescue the loss of RhoU cardiac defect. Finally, we found that Wnt signaling, which has been implicated in atrioventricular canal development (Verhoeven et al., 2011), may regulate the expression of RhoU at the atrioventricular canal. Overall, these findings reveal a cardiac developmental pathway involving RhoU/Arhgef7b/Pak signaling, which helps coordinate cell junction formation between atrioventricular cardiomyocytes to promote cell adhesiveness and cell shapes during cardiac morphogenesis. Failure to properly form these cell adhesions during cardiac development may lead to structural heart defects and mechanistically account for the cellular events that occur in certain human congenital heart diseases.
Collapse
Affiliation(s)
- Michael Dickover
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093-0613J, USA
| | - Jeffrey M Hegarty
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093-0613J, USA
| | - Kim Ly
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093-0613J, USA
| | - Diana Lopez
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093-0613J, USA
| | - Hongbo Yang
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093-0613J, USA
| | - Ruilin Zhang
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093-0613J, USA
| | - Neil Tedeschi
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093-0613J, USA
| | - Tzung K Hsiai
- Department of Medicine, Division of Cardiology, University of Southern California, Los Angeles, CA, USA
| | - Neil C Chi
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093-0613J, USA; Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
76
|
Cardiac regeneration in non-mammalian vertebrates. Exp Cell Res 2014; 321:58-63. [DOI: 10.1016/j.yexcr.2013.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 07/26/2013] [Accepted: 08/01/2013] [Indexed: 01/08/2023]
|
77
|
Yang J, Hartjes KA, Nelson TJ, Xu X. Cessation of contraction induces cardiomyocyte remodeling during zebrafish cardiogenesis. Am J Physiol Heart Circ Physiol 2013; 306:H382-95. [PMID: 24322613 DOI: 10.1152/ajpheart.00721.2013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Contraction regulates heart development via a complex mechanotransduction process controlled by various mechanical forces. Here, we exploit zebrafish embryos as an in vivo animal model to discern the contribution from different mechanical forces and identify the underlying mechanotransductive signaling pathways of cardiogenesis. We treated 2 days postfertilization zebrafish embryos with Blebbistatin, a myosin II inhibitor, to stop cardiac contraction, which induces a response termed cessation of contraction-induced cardiomyocyte (CM) enlargement (CCE). Accompanying the CCE, lateral fusion of myofibrils was attenuated within CMs. The CCE can be blunted by loss of blood in tail-docked zebrafish but not in cloche mutant fish, suggesting that transmural pressure rather than shear stress is accountable for the chamber enlargement. By screening a panel of small molecule inhibitors, our data suggested essential functions of phosphoinositide 3-kinase signaling and protein synthesis in CCE, which are independent of the sarcomere integrity. In summary, we defined a unique CCE response in genetically tractable zebrafish embryos. A panel of assays was established to verify the contribution from extrinsic forces and interrogate underlying signaling pathways.
Collapse
Affiliation(s)
- Jingchun Yang
- Department of Biochemistry and Molecular Biology, Division of Cardiovascular Diseases, Mayo Clinic College of Medicine, Rochester, Minnesota
| | | | | | | |
Collapse
|
78
|
Poon KL, Brand T. The zebrafish model system in cardiovascular research: A tiny fish with mighty prospects. Glob Cardiol Sci Pract 2013; 2013:9-28. [PMID: 24688998 PMCID: PMC3963735 DOI: 10.5339/gcsp.2013.4] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 01/29/2013] [Indexed: 12/26/2022] Open
Affiliation(s)
- Kar Lai Poon
- Harefield Heart Science Centre, National Heart and Lung Institute, Imperial College London, Hill End Road, Harefield, Middlesex, UB9 6JH, United Kingdom
| | - Thomas Brand
- Harefield Heart Science Centre, National Heart and Lung Institute, Imperial College London, Hill End Road, Harefield, Middlesex, UB9 6JH, United Kingdom
| |
Collapse
|
79
|
Lin CY, Huang CC, Wang WD, Hsiao CD, Cheng CF, Wu YT, Lu YF, Hwang SPL. Low temperature mitigates cardia bifida in zebrafish embryos. PLoS One 2013; 8:e69788. [PMID: 23922799 PMCID: PMC3724881 DOI: 10.1371/journal.pone.0069788] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 06/12/2013] [Indexed: 11/18/2022] Open
Abstract
The coordinated migration of bilateral cardiomyocytes and the formation of the cardiac cone are essential for heart tube formation. We investigated gene regulatory mechanisms involved in myocardial migration, and regulation of the timing of cardiac cone formation in zebrafish embryos. Through screening of zebrafish treated with ethylnitrosourea, we isolated a mutant with a hypomorphic allele of mil (s1pr2)/edg5, called s1pr2as10 (as10). Mutant embryos with this allele expressed less mil/edg5 mRNA and exhibited cardia bifida prior to 28 hours post-fertilization. Although the bilateral hearts of the mutants gradually fused together, the resulting formation of two atria and one tightly-packed ventricle failed to support normal blood circulation. Interestingly, cardia bifida of s1pr2as10 embryos could be rescued and normal circulation could be restored by incubating the embryos at low temperature (22.5°C). Rescue was also observed in gata5 and bon cardia bifida morphants raised at 22.5°C. The use of DNA microarrays, digital gene expression analyses, loss-of-function, as well as mRNA and protein rescue experiments, revealed that low temperature mitigates cardia bifida by regulating the expression of genes encoding components of the extracellular matrix (fibronectin 1, tenascin-c, tenascin-w). Furthermore, the addition of N-acetyl cysteine (NAC), a reactive oxygen species (ROS) scavenger, significantly decreased the effect of low temperature on mitigating cardia bifida in s1pr2as10 embryos. Our study reveals that temperature coordinates the development of the heart tube and somitogenesis, and that extracellular matrix genes (fibronectin 1, tenascin-c and tenascin-w) are involved.
Collapse
Affiliation(s)
- Che-Yi Lin
- Institute of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
80
|
8-Oxoguanine DNA glycosylase 1 (ogg1) maintains the function of cardiac progenitor cells during heart formation in zebrafish. Exp Cell Res 2013; 319:2954-63. [PMID: 23892003 DOI: 10.1016/j.yexcr.2013.07.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Revised: 07/08/2013] [Accepted: 07/10/2013] [Indexed: 11/27/2022]
Abstract
Genomic damage may devastate the potential of progenitor cells and consequently impair early organogenesis. We found that ogg1, a key enzyme initiating the base-excision repair, was enriched in the embryonic heart in zebrafish. So far, little is known about DNA repair in cardiogenesis. Here, we addressed the critical role of ogg1 in cardiogenesis for the first time. ogg1 mainly expressed in the anterior lateral plate mesoderm (ALPM), the primary heart tube, and subsequently the embryonic myocardium by in situ hybridisation. Loss of ogg1 resulted in severe cardiac morphogenesis and functional abnormalities, including the short heart length, arrhythmia, decreased cardiomyocytes and nkx2.5(+) cardiac progenitor cells. Moreover, the increased apoptosis and repressed proliferation of progenitor cells caused by ogg1 deficiency might contribute to the heart phenotype. The microarray analysis showed that the expression of genes involved in embryonic heart tube morphogenesis and heart structure were significantly changed due to the lack of ogg1. Among those, foxh1 is an important partner of ogg1 in the cardiac development in response to DNA damage. Our work demonstrates the requirement of ogg1 in cardiac progenitors and heart development in zebrafish. These findings may be helpful for understanding the aetiology of congenital cardiac deficits.
Collapse
|
81
|
Gays D, Santoro MM. The admiR-able advances in cardiovascular biology through the zebrafish model system. Cell Mol Life Sci 2013; 70:2489-503. [PMID: 23069988 PMCID: PMC11113687 DOI: 10.1007/s00018-012-1181-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 09/12/2012] [Accepted: 09/24/2012] [Indexed: 12/30/2022]
Abstract
MicroRNAs are small non-coding RNAs endogenously expressed by all tissues during development and adulthood. They regulate gene expression by controlling the stability of targeted messenger RNA. In cardiovascular tissues microRNAs play a role by modulating essential genes involved in heart and blood vessel development and homeostasis. The zebrafish (Danio rerio) system is a recognized vertebrate model system useful to study cardiovascular biology; recently, it has been used to investigate microRNA functions during natural and pathological states. In this review, we will illustrate the advantages of the zebrafish model in the study of microRNAs in heart and vascular cells, providing an update on recent discoveries using the zebrafish to identify new microRNAs and their targeted genes in cardiovascular tissues. Lastly, we will provide evidence that the zebrafish is an optimal model system to undercover new microRNA functions in vertebrates and to improve microRNA-based therapeutic approaches.
Collapse
Affiliation(s)
- Dafne Gays
- Department of Biology, Biochemistry and Genetics, Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Massimo Mattia Santoro
- Department of Biology, Biochemistry and Genetics, Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Turin, Italy
| |
Collapse
|
82
|
Dyer LA, Patterson C. A novel ex vivo culture method for the embryonic mouse heart. J Vis Exp 2013:e50359. [PMID: 23728379 DOI: 10.3791/50359] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Developmental studies in the mouse are hampered by the inaccessibility of the embryo during gestation. Thus, protocols to isolate and culture individual organs of interest are essential to provide a method of both visualizing changes in development and allowing novel treatment strategies. To promote the long-term culture of the embryonic heart at late stages of gestation, we developed a protocol in which the excised heart is cultured in a semi-solid, dilute Matrigel. This substrate provides enough support to maintain the three-dimensional structure but is flexible enough to allow continued contraction. In brief, hearts are excised from the embryo and placed in a mixture of cold Matrigel diluted 1:1 with growth medium. After the diluted Matrigel solidifies, growth medium is added to the culture dish. Hearts excised as late as embryonic day 16.5 were viable for four days post-dissection. Analysis of the coronary plexus shows that this method does not disrupt coronary vascular development. Thus, we present a novel method for long-term culture of embryonic hearts.
Collapse
Affiliation(s)
- Laura A Dyer
- McAllister Heart Institute, University of North Carolina at Chapel Hill, USA.
| | | |
Collapse
|
83
|
Shimoda H, Isogai S. Immunohistochemical demonstration of lymphatic vessels in adult zebrafish. Acta Histochem Cytochem 2012; 45:335-41. [PMID: 23378677 PMCID: PMC3554784 DOI: 10.1267/ahc.12027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 09/04/2012] [Indexed: 11/22/2022] Open
Abstract
Morphological profiles of lymphatic vessels in adult zebrafish trunk and ovary were studied by immunohistochemistry and electron microscopy. The present immunohistochemistry for Prox1 was successful in demonstrating lymphatic vessels in zebrafish. The zebrafish trunk revealed two types of bilateral longitudinal lymphatic trunks draining lymph centripetally along dorsal aorta and posterior cardinal veins. Large honeycomb lymphatic sinus was further shown around common cardinal veins. In the zebrafish ovary, the lymphatic vessels, comprising endothelial cells only, encompassed arterioles in their lumen. This peculiar structure appeared to be conserved in vertebrates including mammals and might serve for control of blood temperature and tissue homeostasis. The present study is first to delineate lymphatic vessels in adult zebrafish by immunohistochemistry. Our immunohistochemical results showed usefulness of immunostaining for Prox1 not only for demonstration of lymphatic vessels in zebrafish, but also for examination of their function and dynamics in pathophysiological condition.
Collapse
Affiliation(s)
- Hiroshi Shimoda
- Department of Anatomical Science, Graduate School of Medicine, Hirosaki University
- Department of Human Anatomy, Faculty of Medicine, Oita University
| | - Sumio Isogai
- Department of Anatomy, School of Medicine, Iwate Medical University
| |
Collapse
|
84
|
Xia S, Zhu Y, Xu X, Xia W. Computational techniques in zebrafish image processing and analysis. J Neurosci Methods 2012; 213:6-13. [PMID: 23219894 DOI: 10.1016/j.jneumeth.2012.11.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 11/10/2012] [Accepted: 11/19/2012] [Indexed: 12/11/2022]
Abstract
The zebrafish (Danio rerio) has been widely used as a vertebrate animal model in neurobiological. The zebrafish has several unique advantages that make it well suited for live microscopic imaging, including its fast development, large transparent embryos that develop outside the mother, and the availability of a large selection of mutant strains. As the genome of zebrafish has been fully sequenced it is comparatively easier to carry out large scale forward genetic screening in zebrafish to investigate relevant human diseases, from neurological disorders like epilepsy, Alzheimer's disease, and Parkinson's disease to other conditions, such as polycystic kidney disease and cancer. All of these factors contribute to an increasing number of microscopic images of zebrafish that require advanced image processing methods to objectively, quantitatively, and quickly analyze the image dataset. In this review, we discuss the development of image analysis and quantification techniques as applied to zebrafish images, with the emphasis on phenotype evaluation, neuronal structure quantification, vascular structure reconstruction, and behavioral monitoring. Zebrafish image analysis is continually developing, and new types of images generated from a wide variety of biological experiments provide the dataset and foundation for the future development of image processing algorithms.
Collapse
Affiliation(s)
- Shunren Xia
- Key laboratory of Biomedical Engineering, of Ministry of Education Zhejiang University, Hangzhou, China.
| | | | | | | |
Collapse
|
85
|
|