1
|
Aranguren-Abadía L, Yadetie F, Donald CE, Sørhus E, Myklatun LE, Zhang X, Lie KK, Perrichon P, Nakken CL, Durif C, Shema S, Browman HI, Skiftesvik AB, Goksøyr A, Meier S, Karlsen OA. Photo-enhanced toxicity of crude oil on early developmental stages of Atlantic cod (Gadus morhua). THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 807:150697. [PMID: 34610396 DOI: 10.1016/j.scitotenv.2021.150697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/26/2021] [Accepted: 09/26/2021] [Indexed: 06/13/2023]
Abstract
Photo-enhanced toxicity of crude oil is produced by exposure to ultraviolet (UV) radiation. Atlantic cod (Gadus morhua) embryos were exposed to crude oil with and without UV radiation (290-400 nm) from 3 days post fertilization (dpf) until 6 dpf. Embryos from the co-exposure experiment were continually exposed to UV radiation until hatching at 11 dpf. Differences in body burden levels and cyp1a expression in cod embryos were observed between the exposure regimes. High doses of crude oil produced increased mortality in cod co-exposed embryos, as well as craniofacial malformations and heart deformities in larvae from both experiments. A higher number of differentially expressed genes (DEGs) and pathways were revealed in the co-exposure experiment, indicating a photo-enhanced effect of crude oil toxicity. Our results provide mechanistic insights into crude oil and photo-enhanced crude oil toxicity, suggesting that UV radiation increases the toxicity of crude oil in early life stages of Atlantic cod.
Collapse
Affiliation(s)
| | - Fekadu Yadetie
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | | | - Elin Sørhus
- Institute of Marine Research, Bergen, Norway
| | | | - Xiaokang Zhang
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Kai K Lie
- Institute of Marine Research, Bergen, Norway
| | | | | | - Caroline Durif
- Institute of Marine Research, Austevoll Research Station, Storebø, Norway
| | - Steven Shema
- Grótti ehf., Grundarstíg 4, 101 Reykjavík, Iceland
| | - Howard I Browman
- Institute of Marine Research, Austevoll Research Station, Storebø, Norway
| | | | - Anders Goksøyr
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | | | - Odd André Karlsen
- Department of Biological Sciences, University of Bergen, Bergen, Norway.
| |
Collapse
|
2
|
Perumal E, Eswaran S, Parvin R, Balasubramanian S. Mitigation of arsenic induced developmental cardiotoxicity by ferulic acid in zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2021; 244:109021. [PMID: 33631344 DOI: 10.1016/j.cbpc.2021.109021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 02/09/2021] [Accepted: 02/16/2021] [Indexed: 12/29/2022]
Abstract
We investigated whether ferulic acid (FA), a nutraceutical could mitigate the arsenic (As) induced cardiotoxicity. Zebrafish larvae (60 and 72 h post-fertilization [hpf]) were used to study the effect of FA on As at different time points (24 and 48 h after exposure). The FA exposure was given as pre-treatment (60 hpf) and simultaneous treatment (72 hpf) to translate the results for As contaminated areas. To accomplish this, the lethality assay was done, and based on the results, the dosage for As (1 mM) and FA (30 μM) was fixed. The FA intervention (30 μM) as 12 h pre-treatment (60 hpf) and simultaneous treatment along with As (72 hpf) decreased the As content in zebrafish larvae as evidenced by inductively coupled plasma-mass spectrometry. As exposure showed congenital deformities especially cardiac malformations in zebrafish larvae after 24 and 48 h. These teratogenic effects induced by As were reduced by FA supplementation in both groups. Also, o-dianisidine staining demonstrated that As treated larvae encountered abnormal cardiac function with reduced blood circulation, while FA supplementation reversed these effects. The acetylcholinesterase activity, a biomarker of As-induced cardiotoxicity was also found to be decreased in As group, which was rescued by FA. The modulation in the expression of the genes involved in cardiogenesis (nkx2.5, bmp2b, gata4, gata5, myh6, myl7, and tnnt2) further confirmed the ameliorative effect of FA on As induced malformations.
Collapse
Affiliation(s)
- Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India.
| | - Sangavi Eswaran
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | - Reshma Parvin
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | | |
Collapse
|
3
|
Yao Y, Marra AN, Yelon D. Pathways Regulating Establishment and Maintenance of Cardiac Chamber Identity in Zebrafish. J Cardiovasc Dev Dis 2021; 8:13. [PMID: 33572830 PMCID: PMC7912383 DOI: 10.3390/jcdd8020013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 02/07/2023] Open
Abstract
The vertebrate heart is comprised of two types of chambers-ventricles and atria-that have unique morphological and physiological properties. Effective cardiac function depends upon the distinct characteristics of ventricular and atrial cardiomyocytes, raising interest in the genetic pathways that regulate chamber-specific traits. Chamber identity seems to be specified in the early embryo by signals that establish ventricular and atrial progenitor populations and trigger distinct differentiation pathways. Intriguingly, chamber-specific features appear to require active reinforcement, even after myocardial differentiation is underway, suggesting plasticity of chamber identity within the developing heart. Here, we review the utility of the zebrafish as a model organism for studying the mechanisms that establish and maintain cardiac chamber identity. By combining genetic and embryological approaches, work in zebrafish has revealed multiple players with potent influences on chamber fate specification and commitment. Going forward, analysis of cardiomyocyte identity at the single-cell level is likely to yield a high-resolution understanding of the pathways that link the relevant players together, and these insights will have the potential to inform future strategies in cardiac tissue engineering.
Collapse
Affiliation(s)
| | | | - Deborah Yelon
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; (Y.Y.); (A.N.M.)
| |
Collapse
|
4
|
Wu Y, Zhang Y, Chen M, Yang Q, Zhuang S, Lv L, Zuo Z, Wang C. Exposure to low-level metalaxyl impacts the cardiac development and function of zebrafish embryos. J Environ Sci (China) 2019; 85:1-8. [PMID: 31471016 DOI: 10.1016/j.jes.2019.03.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 03/21/2019] [Accepted: 03/21/2019] [Indexed: 06/10/2023]
Abstract
Metalaxyl is an anilide pesticide that is widely used to control plant diseases caused by Peronosporales species. In order to study the toxic effects, zebrafish embryos were exposed to metalaxyl at nominal concentrations of 5, 50 and 500 ng/L for 72 hr, and the cardiac development and functioning of larvae were observed. The results showed that metalaxyl exposure resulted in increased rates of pericardial edema, heart hemorrhage and cardiac malformation. The distance between the sinus venosus and bulbus arteriosus, stroke volume, cardiac output and heart rate were significantly increased in larvae exposed to 50 and 500 ng/L metalaxyl compared to solvent control larvae. Significant upregulation in the transcription of tbx5, gata4 and myh6 was observed in the 50 and 500 ng/L treatments, and that of nkx2.5 and myl7 was observed in the 5, 50 and 500 ng/L groups. These disturbances may be related to cardiac developmental and functional defects in the larvae. The activity of Na+/K+-ATPase and Ca2+-ATPase was significantly increased in zebrafish embryos exposed to 500 ng/L metalaxyl, and the mRNA levels of genes related to ATPase (atp2a11, atp1b2b, and atp1a3b) (in the 50 and 500 ng/L groups) and calcium channels (cacna1ab) (in the 500 ng/L group) were significantly downregulated; these changes might be associated with heart arrhythmia and functional failure.
Collapse
Affiliation(s)
- Yuqiong Wu
- Wuyi University, College of Tea and Food Science, Wuyishan 354300, China.
| | - Ying Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361005, China.
| | - Meng Chen
- Key Laboratory of Ministry of Education for Subtropical Wetland Ecosystem Research, Xiamen University, Xiamen 361005, China
| | - Qihong Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361005, China
| | - Shanshan Zhuang
- Key Laboratory of Ministry of Education for Subtropical Wetland Ecosystem Research, Xiamen University, Xiamen 361005, China
| | - Liangju Lv
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361005, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361005, China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361005, China.
| |
Collapse
|
5
|
Wang W, Wang B, Liu Z, Xia X. Developmental toxicity and alteration of gene expression in zebrafish embryo exposed to 6-benzylaminopurine. CHEMOSPHERE 2019; 233:336-346. [PMID: 31176896 DOI: 10.1016/j.chemosphere.2019.05.261] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 06/09/2023]
Abstract
6-benzylaminopurine (6-BA) is widely used in agriculture and horticulture as plant growth regulator. Its excessive use may pose a potential risk to both environment and human health, which is causing great concern. This study was undertaken to assess the acute developmental toxicity of 6-BA to zebrafish embryos based on OECD protocols and mortality, hatching rate and malformation were investigated. Results showed that the 96 h-LC50 and 96 h- EC50 values were 63.29 mg/L and 41.86 mg/L, respectively. No mortality or teratogenic effects were found at concentrations lower than 10 mg/L 6-BA at concentrations higher than 50 mg/L significantly inhibited hatchability and embryo development, induced serious toxicity characterized by morphologic abnormalities (elongated pericardium, heart and yolk sac edema, spine curvature) and functional failure (slow spontaneous movement and heart rate, growth retardation, yolk sac absorption retention). Moreover, 6-BA-induced apoptosis was observed in embryos by the acridine orange staining and confirmed by the apoptotic-related genes, all of which p53 was significantly up-regulated at concentrations higher than 10 mg/L, bax at concentrations higher than 12.5 mg/L, while bcl2 was down-regulated at concentrations higher than 25 mg/L. As for genes of cardiac development, qPCR results demonstrated that nkx2.5, gata5, and amhc were significantly down-regulated at concentrations higher than 25 mg/L, vmhc and atp2a2a at concentration of 50 mg/L, in contrast, hand2 was up-regulated at concentration of 50 mg/L. Our data indicate that 6-BA induces a dose-dependent toxicity resulting in apoptosis through the involvement of p53-dependent pathways and hindering normal heart development in zebrafish embryos.
Collapse
Affiliation(s)
- Weixiang Wang
- School of Food and Bioengineering, Xihua University, Chengdu, PR China.
| | - Binrong Wang
- School of Food and Bioengineering, Xihua University, Chengdu, PR China
| | - Zihao Liu
- School of Food and Bioengineering, Xihua University, Chengdu, PR China
| | - Xuemei Xia
- School of Food and Bioengineering, Xihua University, Chengdu, PR China
| |
Collapse
|
6
|
Wu Y, Yang Q, Chen M, Zhang Y, Zuo Z, Wang C. Fenbuconazole exposure impacts the development of zebrafish embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 158:293-299. [PMID: 29715634 DOI: 10.1016/j.ecoenv.2018.04.048] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/16/2018] [Accepted: 04/21/2018] [Indexed: 06/08/2023]
Abstract
Fenbuconazole (FBZ), a triazole-containing fungicide, is widely used in agriculture and horticulture. In the present study, the development and cardiac functioning were observed and determined in zebrafish embryos exposed to FBZ at 5, 50 and 500 ng/L nominal concentrations for 72 h. The results showed that 500 ng/L FBZ significantly increased pericardial edema rate, spine curvature rate, disturbed cardiac function, and led a shortened lower jaw. The transcription of genes such as tbx5, nkx2.5, tnnt2, gata4, bmp2b, myl7 was altered, which might be responsible for the cardiac developmental and functioning defects in the larvae. The deformation in bone development might be related with the impaired transcription levels of shh and bmp2b. The transcription of cyp26a1 (encoding retinoic acid metabolism enzyme) was significantly up-regulated in the 500 ng/L group, which might be a reason causing the teratogenic effect of FBZ. These results suggest that FBZ could have toxic effects on embryonic development, which should be considered in the risk evaluation of FBZ application.
Collapse
Affiliation(s)
- Yuqiong Wu
- Wuyi University, College of Tea and Food Science, Wuyishan, Fujian 354300, China
| | - Qihong Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Meng Chen
- Key Laboratory of Ministry of Education for Subtropical Wetland Ecosystem Research, Xiamen University, Xiamen, Fujian 361005, China
| | - Ying Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China.
| |
Collapse
|
7
|
Fukui H, Miyazaki T, Chow RWY, Ishikawa H, Nakajima H, Vermot J, Mochizuki N. Hippo signaling determines the number of venous pole cells that originate from the anterior lateral plate mesoderm in zebrafish. eLife 2018; 7:29106. [PMID: 29809141 PMCID: PMC5995544 DOI: 10.7554/elife.29106] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 05/26/2018] [Indexed: 12/11/2022] Open
Abstract
The differentiation of the lateral plate mesoderm cells into heart field cells constitutes a critical step in the development of cardiac tissue and the genesis of functional cardiomyocytes. Hippo signaling controls cardiomyocyte proliferation, but the role of Hippo signaling during early cardiogenesis remains unclear. Here, we show that Hippo signaling regulates atrial cell number by specifying the developmental potential of cells within the anterior lateral plate mesoderm (ALPM), which are incorporated into the venous pole of the heart tube and ultimately into the atrium of the heart. We demonstrate that Hippo signaling acts through large tumor suppressor kinase 1/2 to modulate BMP signaling and the expression of hand2, a key transcription factor that is involved in the differentiation of atrial cardiomyocytes. Collectively, these results demonstrate that Hippo signaling defines venous pole cardiomyocyte number by modulating both the number and the identity of the ALPM cells that will populate the atrium of the heart.
Collapse
Affiliation(s)
- Hajime Fukui
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan.,University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Takahiro Miyazaki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Renee Wei-Yan Chow
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Hiroyuki Ishikawa
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Hiroyuki Nakajima
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan.,AMED-Core Research for Evolutional Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| |
Collapse
|
8
|
Wu Y, Zuo Z, Chen M, Zhou Y, Yang Q, Zhuang S, Wang C. The developmental effects of low-level procymidone towards zebrafish embryos and involved mechanism. CHEMOSPHERE 2018; 193:928-935. [PMID: 29874768 DOI: 10.1016/j.chemosphere.2017.11.105] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 06/08/2023]
Abstract
Procymidone (PCM), a dicarboximide fungicide, is widely used in agriculture to control plant diseases. In the present study, zebrafish embryos were exposed to PCM at 0, 10, 100 and 1000 ng/L for 72 h, the development and cardiac functioning of larvae were observed and determined. The results showed that hatching rate was significantly decreased in the 1000 ng/L treatment, and pericardial edema rate and spine curvature rate were significantly increased in the 100 and 1000 ng/L groups. The PCM-treated larvae exhibited an increased heart rate as well as arrhythmia, and shortened low jaw. The transcription levels of cardiac development-related genes tbx5, nkx2.5, tnnt2, gata4, myh6, myl7, cdh2, ryr2 were altered, which might be responsible for the cardiac developmental and functioning defects in the larvae. The deformation in bone development might be related with the impaired transcription levels of ihh, shh, bmp2b, bmp4, gh, igf1, sox9, gli2. The activities of Na+/K+-ATPase and Ca2+-ATPase were significantly inhibited by 100 ng/L and 1000 ng/L PCM exposure, which might be a cause for the occurrence of pericardial edema and skeletal deformation. The results of this study will be helpful in evaluating the potential threat of PCM to fish population in the aquatic environment.
Collapse
Affiliation(s)
- Yuqiong Wu
- Wuyi University, College of Tea and Food Science, Wuyishan, Fujian 354300, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Meng Chen
- Key Laboratory of the Coastal and Wetland Ecosystems (Xiamen University), Ministry of Education, China
| | - Yixi Zhou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Qihong Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Shanshan Zhuang
- Key Laboratory of the Coastal and Wetland Ecosystems (Xiamen University), Ministry of Education, China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China.
| |
Collapse
|
9
|
Fukui H, Chiba A, Miyazaki T, Takano H, Ishikawa H, Omori T, Mochiuzki N. Spatial Allocation and Specification of Cardiomyocytes during Zebrafish Embryogenesis. Korean Circ J 2017; 47:160-167. [PMID: 28382067 PMCID: PMC5378018 DOI: 10.4070/kcj.2016.0280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 10/05/2016] [Accepted: 10/13/2016] [Indexed: 12/21/2022] Open
Abstract
Incomplete development and severe malformation of the heart result in miscarriage of embryos because of its malfunction as a pump for circulation. During cardiogenesis, development of the heart is precisely coordinated by the genetically-primed program that is revealed by the sequential expression of transcription factors. It is important to investigate how spatial allocation of the heart containing cardiomyocytes and other mesoderm-derived cells is determined. In addition, the molecular mechanism underlying cardiomyocyte differentiation still remains elusive. The location of ectoderm-, mesoderm-, and endoderm-derived organs is determined by their initial allocation and subsequent mutual cell-cell interactions or paracrine-based regulation. In the present work, we provide an overview of cardiac development controlled by the germ layers and discuss the points that should be uncovered in future for understanding cardiogenesis.
Collapse
Affiliation(s)
- Hajime Fukui
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Ayano Chiba
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Takahiro Miyazaki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Haruko Takano
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Hiroyuki Ishikawa
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Toyonori Omori
- Management office, National Center for Child Health and Development, Tokyo, Japan
| | - Naoki Mochiuzki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan.; AMED-CREST, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| |
Collapse
|
10
|
Wen B, Yuan H, Liu X, Wang H, Chen S, Chen Z, de The H, Zhou J, Zhu J. GATA5 SUMOylation is indispensable for zebrafish cardiac development. Biochim Biophys Acta Gen Subj 2017; 1861:1691-1701. [PMID: 28285006 DOI: 10.1016/j.bbagen.2017.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 01/07/2017] [Accepted: 03/07/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND SUMOylation is a critical regulatory protein modification in eukaryotic cells and plays a pivotal role in cardiac development and disease. Several cardiac transcription factors are modified by SUMO, but little is known about the impact of SUMOylation on their function during cardiac development. METHODS We used a zebrafish model to address the impact of SUMOylation on GATA5, an essential transcription factor in zebrafish cardiac development. GATA5 SUMOylation was probed by western blot, the subcellular localization and transcriptional activity of GATA5 mutants were examined by immunostaining and luciferase reporter assay. The in vivo function of GATA5 SUMOylation was evaluated by gata5 mutants mRNA microinjection and in situ hybridization in gata5 morphants and ubc9 mutants. RESULTS Firstly, we identified GATA5 as a SUMO substrate, and lysine 324 (K324) and lysine 360 (K360) as two major modification sites. Conversion of lysine to arginine at these two sites did not affect subcellular localization, but did affect the transcriptional activity of GATA5. Secondly, in vivo experiments demonstrated that the wild type (WT) and K324R mutant of gata5 could rescue impaired cardiac precursor differentiation, while the K360R mutant of gata5 drastically lost this potency in gata5 morphant. Furthermore, in SUMOylation-deficient ubc9 mutants, the abnormal expression pattern displayed by the early markers of cardiac development (nkx2.5 and mef2cb) could be restored using a sumo-gata5 fusion, but not with a WT gata5. CONCLUSION GATA5 SUMOylation is indispensable for early zebrafish cardiac development. GENERAL SIGNIFICANCE Our studies highlight the potential importance of transcription factor SUMOylation in cardiac development.
Collapse
Affiliation(s)
- Bin Wen
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Yuan
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohui Liu
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haihong Wang
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Saijuan Chen
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhu Chen
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hugues de The
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Université de Paris 7/INSERM/CNRS UMR 944/7212, Equipe Labellisée No. 11 Ligue Nationale Contre le Cancer, Hôpital St. Louis, Paris, France
| | - Jun Zhou
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jun Zhu
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Université de Paris 7/INSERM/CNRS UMR 944/7212, Equipe Labellisée No. 11 Ligue Nationale Contre le Cancer, Hôpital St. Louis, Paris, France.
| |
Collapse
|
11
|
Ectopic expression of Cripto-1 in transgenic mouse embryos causes hemorrhages, fatal cardiac defects and embryonic lethality. Sci Rep 2016; 6:34501. [PMID: 27687577 PMCID: PMC5043281 DOI: 10.1038/srep34501] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 09/15/2016] [Indexed: 12/23/2022] Open
Abstract
Targeted disruption of Cripto-1 in mice caused embryonic lethality at E7.5, whereas we unexpectedly found that ectopic Cripto-1 expression in mouse embryos also led to embryonic lethality, which prompted us to characterize the causes and mechanisms underlying embryonic death due to ectopic Cripto-1 expression. RCLG/EIIa-Cre embryos displayed complex phenotypes between embryonic day 14.5 (E14.5) and E17.5, including fatal hemorrhages (E14.5-E15.5), embryo resorption (E14.5-E17.5), pale body surface (E14.5-E16.5) and no abnormal appearance (E14.5-E16.5). Macroscopic and histological examination revealed that ectopic expression of Cripto-1 transgene in RCLG/EIIa-Cre embryos resulted in lethal cardiac defects, as evidenced by cardiac malformations, myocardial thinning, failed assembly of striated myofibrils and lack of heartbeat. In addition, Cripto-1 transgene activation beginning after E8.5 also caused the aforementioned lethal cardiac defects in mouse embryos. Furthermore, ectopic Cripto-1 expression in embryonic hearts reduced the expression of cardiac transcription factors, which is at least partially responsible for the aforementioned lethal cardiac defects. Our results suggest that hemorrhages and cardiac abnormalities are two important lethal factors in Cripto-1 transgenic mice. Taken together, these findings are the first to demonstrate that sustained Cripto-1 transgene expression after E11.5 causes fatal hemorrhages and lethal cardiac defects, leading to embryonic death at E14.5-17.5.
Collapse
|
12
|
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
|
13
|
Yang S, Wang S, Sun F, Zhang M, Wu F, Xu F, Ding Z. Protective effects of puerarin against tetrabromobisphenol a-induced apoptosis and cardiac developmental toxicity in zebrafish embryo-larvae. ENVIRONMENTAL TOXICOLOGY 2015; 30:1014-1023. [PMID: 24596333 DOI: 10.1002/tox.21975] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 02/10/2014] [Accepted: 02/23/2014] [Indexed: 06/03/2023]
Abstract
Tetrabromobisphenol A (TBBPA), a brominated flame retardant, is detected commonly in aquatic environments, where it is thought to be highly toxic to the development of aquatic life. In this study, zebrafish embryos and larvae were used to investigate the protective effects of puerarin after exposure to TBBPA. Malformation, blood flow disorders, pericardial edema, and spawn coagulation rates increased, whereas survival decreased significantly after exposure to 0.5 and 1.0 mg L(-1) TBBPA. The measured indices of morphological toxicity improved after treatment with puerarin. TBBPA also induced reactive oxygen species (ROS) production in a dose-dependent manner. Acridine orange staining results revealed that TBBPA exposure caused cardiomyocyte apoptosis and induced the expression of three proapoptotic genes: P53, Bax, and Caspase9. In contrast, the expression of the antiapoptotic gene Bcl2 was down-regulated. When genes related to cardiac development were assessed, the expression of Tbx1, Raldh2, and Bmp2b changed after exposure to the combination of TBBPA and puerarin. These results suggest that TBBPA induces cardiomyocyte apoptosis and ROS production, resulting in cardiac developmental toxicity in zebrafish embryos or larvae. Therefore, puerarin regulates the expression of cardiac developmental genes, such as Tbx1, Bmp2b, and Raldh2 by inhibiting ROS production, and subsequently modulates cardiac development after the exposure of zebrafish larvae to TBBPA.
Collapse
Affiliation(s)
- Suwen Yang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Shengrui Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Fengchao Sun
- Department of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mengmeng Zhang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Fengchang Wu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Fanfan Xu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Zhishan Ding
- Department of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| |
Collapse
|
14
|
Xin Q, Rotchell JM, Cheng J, Yi J, Zhang Q. Silver nanoparticles affect the neural development of zebrafish embryos. J Appl Toxicol 2015; 35:1481-92. [PMID: 25976698 DOI: 10.1002/jat.3164] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 02/28/2015] [Accepted: 03/18/2015] [Indexed: 01/04/2023]
Abstract
Silver nanoparticles (AgNPs) have been widely used in commercial products. This study aims to understand the impact of AgNPs on the early developmental stages in zebrafish (Danio rerio) embryos. Embryos were exposed to two sizes of AgNPs at three dose levels, as well to free Ag(+) ions, for a range of 4-96 h post-fertilization (hpf). The acute exposure study showed that exposure to AgNPs affected the neurological development, and the exposed embryos exhibited anomalies such as small head with hypoplastic hindbrain, small eye and cardiac defects. At the molecular level, AgNPs altered the expression profiles of neural development-related genes (gfap, huC and ngn1), metal-sensitive metallothioneins and ABCC genes in exposed embryos. The expression of AhR2 and Cyp1A, which are usually considered to mediate polycyclic aromatic hydrocarbon toxicity, were also significantly changed. A size-dependent uptake of AgNPs was observed, whereby 4 nm AgNPs were more efficiently taken up compared with the 10 nm-sized particles. Importantly, the head area accumulated AgNPs more efficiently than the trunk area of exposed zebrafish embryos. No free Ag(+) ions, which can be potentially released from the AgNP solutions, were detected. This study suggests that AgNPs could affect the neural development of zebrafish embryos, and the toxicity of AgNPs may be partially attributed to the comparatively higher uptake in the head area. These results indicate the potential neurotoxicity of AgNPs and could be extended to other aquatic organisms.
Collapse
Affiliation(s)
- Qi Xin
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai, 200062, China.,City University of Hong Kong Shenzhen Research Institute, Shenzhen, 518057, China
| | - Jeanette M Rotchell
- School of Biological, Biomedical & Environmental Sciences, University of Hull, Cottingham Road, Hull, HU6 7RX, UK
| | - Jinping Cheng
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai, 200062, China.,City University of Hong Kong Shenzhen Research Institute, Shenzhen, 518057, China.,Environmental Science Programs, School of Science, Hong Kong University of Science and Technology, Clear Water bay, Kowloon, Hong Kong, China
| | - Jun Yi
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai, 200062, China
| | - Qiang Zhang
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai, 200062, China
| |
Collapse
|
15
|
Si L, Shi J, Gao W, Zheng M, Liu L, Zhu J, Tian J. Smad4 mediated BMP2 signal is essential for the regulation of GATA4 and Nkx2.5 by affecting the histone H3 acetylation in H9c2 cells. Biochem Biophys Res Commun 2014; 450:81-6. [PMID: 24866243 DOI: 10.1016/j.bbrc.2014.05.068] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 05/17/2014] [Indexed: 02/01/2023]
Abstract
BMP2 signaling pathway plays critical roles during heart development, Smad4 encodes the only common Smad protein in mammals, which is a pivotal nuclear mediator. Our previous studies showed that BMP2 enhanced the expression of cardiac transcription factors in part by increasing histone H3 acetylation. In the present study, we tested the hypothesis that Smad4 mediated BMP2 signaling pathway is essential for the expression of cardiac core transcription factors by affecting the histone H3 acetylation. We successfully constructed a lentivirus-mediated short hairpin RNA interference vector targeting Smad4 (Lv-Smad4) in rat H9c2 embryonic cardiac myocytes (H9c2 cells) and demonstrated that it suppressed the expression of the Smad4 gene. Cultured H9c2 cells were transfected with recombinant adenoviruses expressing human BMP2 (AdBMP2) with or without Lv-Smad4. Quantitative real-time RT-PCR analysis showed that knocking down of Smad4 substantially inhibited both AdBMP2-induced and basal expression levels of cardiac transcription factors GATA4 and Nkx2.5, but not MEF2c and Tbx5. Similarly, chromatin immunoprecipitation (ChIP) analysis showed that knocking down of Smad4 inhibited both AdBMP2-induced and basal histone H3 acetylation levels in the promoter regions of GATA4 and Nkx2.5, but not of Tbx5 and MEF2c. In addition, Lv-Smad4 selectively suppressed AdBMP2-induced expression of HAT p300, but not of HAT GCN5 in H9c2 cells. The data indicated that inhibition of Smad4 diminished both AdBMP2 induced and basal histone acetylation levels in the promoter regions of GATA4 and Nkx2.5, suggesting that Smad4 mediated BMP2 signaling pathway was essential for the regulation of GATA4 and Nkx2.5 by affecting the histone H3 acetylation in H9c2 cells.
Collapse
Affiliation(s)
- Lina Si
- Heart Centre, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing 400014, PR China
| | - Jin Shi
- Heart Centre, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing 400014, PR China
| | - Wenqun Gao
- Heart Centre, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing 400014, PR China
| | - Min Zheng
- Heart Centre, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing 400014, PR China
| | - Lingjuan Liu
- Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing 400014, PR China
| | - Jing Zhu
- Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing 400014, PR China
| | - Jie Tian
- Heart Centre, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing 400014, PR China.
| |
Collapse
|
16
|
Extraembryonic Signals under the Control of MGA, Max, and Smad4 Are Required for Dorsoventral Patterning. Dev Cell 2014; 28:322-34. [DOI: 10.1016/j.devcel.2014.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 11/13/2013] [Accepted: 01/07/2014] [Indexed: 11/19/2022]
|
17
|
Wilkinson RN, Jopling C, van Eeden FJM. Zebrafish as a model of cardiac disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 124:65-91. [PMID: 24751427 DOI: 10.1016/b978-0-12-386930-2.00004-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The zebrafish has been rapidly adopted as a model for cardiac development and disease. The transparency of the embryo, its limited requirement for active oxygen delivery, and ease of use in genetic manipulations and chemical exposure have made it a powerful alternative to rodents. Novel technologies like TALEN/CRISPR-mediated genome engineering and advanced imaging methods will only accelerate its use. Here, we give an overview of heart development and function in the fish and highlight a number of areas where it is most actively contributing to the understanding of cardiac development and disease. We also review the current state of research on a feature that we only could wish to be conserved between fish and human; cardiac regeneration.
Collapse
Affiliation(s)
- Robert N Wilkinson
- Department of Cardiovascular Science, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Chris Jopling
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Département de Physiologie, Labex Ion Channel Science and Therapeutics, Montpellier, France; INSERM, U661, Montpellier, France; Universités de Montpellier 1&2, UMR-5203, Montpellier, France
| | - Fredericus J M van Eeden
- MRC Centre for Biomedical Genetics, Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
18
|
Targoff KL, Colombo S, George V, Schell T, Kim SH, Solnica-Krezel L, Yelon D. Nkx genes are essential for maintenance of ventricular identity. Development 2013; 140:4203-13. [PMID: 24026123 DOI: 10.1242/dev.095562] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Establishment of specific characteristics of each embryonic cardiac chamber is crucial for development of a fully functional adult heart. Despite the importance of defining and maintaining unique features in ventricular and atrial cardiomyocytes, the regulatory mechanisms guiding these processes are poorly understood. Here, we show that the homeodomain transcription factors Nkx2.5 and Nkx2.7 are necessary to sustain ventricular chamber attributes through repression of atrial chamber identity. Mutation of nkx2.5 in zebrafish yields embryos with diminutive ventricular and bulbous atrial chambers. These chamber deformities emerge gradually during development, with a severe collapse in the number of ventricular cardiomyocytes and an accumulation of excess atrial cardiomyocytes as the heart matures. Removal of nkx2.7 function from nkx2.5 mutants exacerbates the loss of ventricular cells and the gain of atrial cells. Moreover, in these Nkx-deficient embryos, expression of vmhc, a ventricular gene, fades, whereas expression of amhc, an atrial gene, expands. Cell-labeling experiments suggest that ventricular cardiomyocytes can transform into atrial cardiomyocytes in the absence of Nkx gene function. Through suggestion of transdifferentiation from ventricular to atrial fate, our data reveal a pivotal role for Nkx genes in maintaining ventricular identity and highlight remarkable plasticity in differentiated myocardium. Thus, our results are relevant to the etiologies of fetal and neonatal cardiac pathology and could direct future innovations in cardiac regenerative medicine.
Collapse
Affiliation(s)
- Kimara L Targoff
- Developmental Genetics Program and Department of Cell Biology, Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
p300-Mediated Histone Acetylation is Essential for the Regulation of GATA4 and MEF2C by BMP2 in H9c2 Cells. Cardiovasc Toxicol 2013; 13:316-22. [DOI: 10.1007/s12012-013-9212-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
20
|
Staudt D, Stainier D. Uncovering the molecular and cellular mechanisms of heart development using the zebrafish. Annu Rev Genet 2012; 46:397-418. [PMID: 22974299 DOI: 10.1146/annurev-genet-110711-155646] [Citation(s) in RCA: 205] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Over the past 20 years, the zebrafish has emerged as a powerful model organism for studying cardiac development. Its ability to survive without an active circulation and amenability to forward genetics has led to the identification of numerous mutants whose study has helped elucidate new mechanisms in cardiac development. Furthermore, its transparent, externally developing embryos have allowed detailed cellular analyses of heart development. In this review, we discuss the molecular and cellular processes involved in zebrafish heart development from progenitor specification to development of the valve and the conduction system. We focus on imaging studies that have uncovered the cellular bases of heart development and on zebrafish mutants with cardiac abnormalities whose study has revealed novel molecular pathways in cardiac cell specification and tissue morphogenesis.
Collapse
Affiliation(s)
- David Staudt
- Department of Biochemistry and Biophysics, University of California, San Francisco, California 94158, USA
| | | |
Collapse
|
21
|
Zhen YS, Wu Q, Xiao CL, Chang NN, Wang X, Lei L, Zhu X, Xiong JW. Overlapping cardiac programs in heart development and regeneration. J Genet Genomics 2012; 39:443-9. [PMID: 23021544 DOI: 10.1016/j.jgg.2012.07.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Revised: 07/02/2012] [Accepted: 07/07/2012] [Indexed: 02/03/2023]
Abstract
Gaining cellular and molecular insights into heart development and regeneration will likely provide new therapeutic targets and opportunities for cardiac regenerative medicine, one of the most urgent clinical needs for heart failure. Here we present a review on zebrafish heart development and regeneration, with a particular focus on early cardiac progenitor development and their contribution to building embryonic heart, as well as cellular and molecular programs in adult zebrafish heart regeneration. We attempt to emphasize that the signaling pathways shaping cardiac progenitors in heart development may also be redeployed during the progress of adult heart regeneration. A brief perspective highlights several important and promising research areas in this exciting field.
Collapse
Affiliation(s)
- Yi-Song Zhen
- Institute of Molecular Medicine, Peking University, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Tu S, Chi NC. Zebrafish models in cardiac development and congenital heart birth defects. Differentiation 2012; 84:4-16. [PMID: 22704690 DOI: 10.1016/j.diff.2012.05.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 05/02/2012] [Accepted: 05/21/2012] [Indexed: 12/31/2022]
Abstract
The zebrafish has become an ideal vertebrate animal system for investigating cardiac development due to its genetic tractability, external fertilization, early optical clarity and ability to survive without a functional cardiovascular system during development. In particular, recent advances in imaging techniques and the creation of zebrafish transgenics now permit the in vivo analysis of the dynamic cellular events that transpire during cardiac morphogenesis. As a result, the combination of these salient features provides detailed insight as to how specific genes may influence cardiac development at the cellular level. In this review, we will highlight how the zebrafish has been utilized to elucidate not only the underlying mechanisms of cardiac development and human congenital heart diseases (CHDs), but also potential pathways that may modulate cardiac regeneration. Thus, we have organized this review based on the major categories of CHDs-structural heart, functional heart, and vascular/great vessel defects, and will conclude with how the zebrafish may be further used to contribute to our understanding of specific human CHDs in the future.
Collapse
Affiliation(s)
- Shu Tu
- Department of Medicine, Division of Cardiology, University of California, San Diego, CA 92093-0613J, USA
| | | |
Collapse
|
23
|
Zhang Y, Wang C, Huang L, Chen R, Chen Y, Zuo Z. Low-level pyrene exposure causes cardiac toxicity in zebrafish (Danio rerio) embryos. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2012; 114-115:119-24. [PMID: 22446823 DOI: 10.1016/j.aquatox.2012.02.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Revised: 02/17/2012] [Accepted: 02/21/2012] [Indexed: 05/06/2023]
Abstract
It is widely accepted that the most abundant polycyclic aromatic hydrocarbons (PAHs) in weathered crude oils is cardiotoxic. Although PAHs toxic endpoints show strong correlation with the aryl hydrocarbon receptor (AhR), a ligand activated transcription factor, and is thought to be a potent inducer of cytochrome P4501A, the action mechanism of PAHs on vertebrate cardiovascular development and disease is unclear. Herein, we address the cardiac developmental effects of exposure to the weak AhR agonist pyrene on the early life-stages of zebrafish. Embryos were exposed to 0, 0.05, 0.5, 5, and 50 nmol/L pyrene up to 72h post-fertilization (hpf). Pyrene-treated embryos showed dose-dependent heart abnormalities, such as pericardial edema and cardiac looping defects. Changes in AhR1a, AhR1b, AhR2, and Cyp1A expression were assessed by real-time RT-PCR. The results showed that low-level pyrene failed to alter these genes expression. However, the homeodomain transcription factor Nkx2.5, which plays an essential role in the development of the cardiovascular system, was down-regulated in a dose-dependent manner by pyrene exposure. The bone morphogenetic protein 2b (Bmp2b), which has been identified as the upstream gene of Nkx2.5, also was inhibited in a dose-dependent manner after treatment with pyrene. Taken together, these data indicated that embryonic exposure of zebrafish to low-level environmental pyrene disrupt normal cardiac development and alter expression of defective cardiac differentiation related genes.
Collapse
Affiliation(s)
- Youyu Zhang
- Key Laboratory of Ministry of Education for Subtropical Wetland Ecosystem Research, School of Life Sciences, Xiamen University, Xiamen, China
| | | | | | | | | | | |
Collapse
|
24
|
Abstract
Heart development is a complex process that involves cell specification and differentiation, as well as elaborate tissue morphogenesis and remodeling, to generate a functional organ. The zebrafish has emerged as a powerful model system to unravel the basic genetic, molecular, and cellular mechanisms of cardiac development and function. We summarize and discuss recent discoveries on early cardiac specification and the identification of the second heart field in zebrafish. In addition to the inductive signals regulating cardiac specification, these studies have shown that heart development also requires a repressive mechanism imposed by retinoic acid signaling to select cardiac progenitors from a multipotent population. Another recent advance in the study of early zebrafish cardiac development is the identification of the second heart field. These studies suggest that the molecular mechanisms that regulate the second heart field development are conserved between zebrafish and other vertebrates including mammals and provide insight into the evolution of the second heart field and its derivatives.
Collapse
Affiliation(s)
- Jiandong Liu
- Department of Biochemistry and Biophysics, University of California, San Francisco, USA.
| | | |
Collapse
|
25
|
Paskaradevan S, Scott IC. The Aplnr GPCR regulates myocardial progenitor development via a novel cell-non-autonomous, Gα(i/o) protein-independent pathway. Biol Open 2012; 1:275-85. [PMID: 23213418 PMCID: PMC3507289 DOI: 10.1242/bio.2012380] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Myocardial progenitor development involves the migration of cells to the anterior lateral plate mesoderm (ALPM) where they are exposed to the necessary signals for heart development to proceed. Whether the arrival of cells to this location is sufficient, or whether earlier signaling events are required, for progenitor development is poorly understood. Here we demonstrate that in the absence of Aplnr signaling, cells fail to migrate to the heart-forming region of the ALPM. Our work uncovers a previously uncharacterized cell-non-autonomous function for Aplnr signaling in cardiac development. Furthermore, we show that both the single known Aplnr ligand, Apelin, and the canonical Gαi/o proteins that signal downstream of Aplnr are dispensable for Aplnr function in the context of myocardial progenitor development. This novel Aplnr signal can be substituted for by activation of Gata5/Smarcd3 in myocardial progenitors, suggesting a novel mechanism for Aplnr signaling in the establishment of a niche required for the proper migration/development of myocardial progenitor cells.
Collapse
Affiliation(s)
- Sivani Paskaradevan
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 555 University Avenue , Toronto, ON M5G 1X8 , Canada ; Department of Molecular Genetics, University of Toronto , Toronto, ON M5S 1A8 , Canada
| | | |
Collapse
|
26
|
Abstract
Ever increasing advances are being made in our quest to understand what it takes to direct pluripotent precursor cells to adopt a specific developmental fate. Eventually, the obvious goal is that targeted manipulation of these precursor cells will result in an efficient and reliable production of tissue-specific cells, which can be safely employed for therapeutic purposes. We have gained an incredible insight as to which molecular pathways are involved in governing neural, skeletal and cardiac muscle fate decisions. However, we still face the challenge of how to direct, for example, a cardiac fate in stem cells in the amounts needed to be employed for regenerative means. Equally importantly, we need to resolve critical questions such as: can the in vitro generated cardiomyocytes actually functionally replace damaged heart tissue? Here I will provide an overview of the molecules and signalling pathways that have first been demonstrated in embryological studies to function in cardiogenesis, and summarize how this knowledge is being applied to differentiate mouse and human embryonic stem cells into cardiomyocytes.
Collapse
Affiliation(s)
- Petra Pandur
- Universität Ulm, Abt. Biochemie, Albert-Einstein-Allee 11, 89081 Ulm, Germany.
| |
Collapse
|
27
|
Ni TT, Rellinger EJ, Mukherjee A, Stephens L, Thorne CA, Kim K, Hu J, Xie S, Lee E, Marnett L, Hatzopoulos AK, Zhong TP. Discovering small molecules that promote cardiomyocyte generation by modulating Wnt signaling. CHEMISTRY & BIOLOGY 2011; 18:1658-68. [PMID: 22195568 PMCID: PMC3645312 DOI: 10.1016/j.chembiol.2011.09.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 09/18/2011] [Accepted: 09/19/2011] [Indexed: 01/12/2023]
Abstract
We have developed a robust in vivo small-molecule screen that modulates heart size and cardiomyocyte generation in zebrafish. Three structurally related compounds (Cardionogen-1 to Cardionogen-3) identified from our screen enlarge the size of the developing heart via myocardial hyperplasia. Increased cardiomyocyte number in Cardionogen-treated embryos is due to expansion of cardiac progenitor cells. In zebrafish embryos and murine embryonic stem (ES) cells, Cardionogen treatment promotes cardiogenesis during and after gastrulation, whereas it inhibits heart formation before gastrulation. Cardionogen-induced effects can be antagonized by increasing Wnt/β-catenin signaling activity. We demonstrate that Cardionogen inhibits Wnt/β-catenin-dependent transcription in murine ES cells and zebrafish embryos. Cardionogen can rescue Wnt8-induced cardiomyocyte deficiency and heart-specific phenotypes during development. These findings demonstrate that in vivo small-molecule screens targeting heart size can reveal compounds with cardiomyogenic effects and identify underlying target pathways.
Collapse
Affiliation(s)
- Terri T. Ni
- State Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 20043, China
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Eric J. Rellinger
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Amrita Mukherjee
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Lauren Stephens
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Cutris A Thorne
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Kwangho Kim
- Department of Chemistry, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Jiangyong Hu
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Shuying Xie
- State Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 20043, China
| | - Ethan Lee
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Larry Marnett
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Antonis K. Hatzopoulos
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Tao P. Zhong
- State Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 20043, China
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| |
Collapse
|
28
|
Farina A, D'Aniello C, Severino V, Hochstrasser DF, Parente A, Minchiotti G, Chambery A. Temporal proteomic profiling of embryonic stem cell secretome during cardiac and neural differentiation. Proteomics 2011; 11:3972-82. [DOI: 10.1002/pmic.201100063] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 05/10/2011] [Accepted: 06/08/2011] [Indexed: 11/08/2022]
|
29
|
Abstract
Over the last decade, the zebrafish has entered the field of cardiovascular research as a new model organism. This is largely due to a number of highly successful small- and large-scale forward genetic screens, which have led to the identification of zebrafish mutants with cardiovascular defects. Genetic mapping and identification of the affected genes have resulted in novel insights into the molecular regulation of vertebrate cardiac development. More recently, the zebrafish has become an attractive model to study the effect of genetic variations identified in patients with cardiovascular defects by candidate gene or whole-genome-association studies. Thanks to an almost entirely sequenced genome and high conservation of gene function compared with humans, the zebrafish has proved highly informative to express and study human disease-related gene variants, providing novel insights into human cardiovascular disease mechanisms, and highlighting the suitability of the zebrafish as an excellent model to study human cardiovascular diseases. In this review, I discuss recent discoveries in the field of cardiac development and specific cases in which the zebrafish has been used to model human congenital and acquired cardiac diseases.
Collapse
Affiliation(s)
- Jeroen Bakkers
- Hubrecht Institute-KNAW & University Medical Center Utrecht, Interuniversity Cardiology Institute of The Netherlands, 3584 CT Utrecht, The Netherlands.
| |
Collapse
|
30
|
Wang J, Greene SB, Martin JF. BMP signaling in congenital heart disease: new developments and future directions. ACTA ACUST UNITED AC 2011; 91:441-8. [PMID: 21384533 DOI: 10.1002/bdra.20785] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 12/22/2010] [Accepted: 01/03/2011] [Indexed: 01/07/2023]
Abstract
Congenital heart malformations are the most common of all congenital human birth anomalies. During the past decade, research with zebrafish, chick, and mouse models have elucidated many fundamental genetic pathways that govern early cardiac patterning and differentiation. This review highlights the roles of the bone morphogenetic protein (BMP) signaling pathway in cardiogenesis and how defective BMP signals can disrupt the intricate steps of cardiac formation and cause congenital heart defects.
Collapse
Affiliation(s)
- Jun Wang
- Institute of Biosciences and Technology, Texas A&M System Health Science Center, 2121 W. Holcombe Blvd., Houston, TX 77030, USA
| | | | | |
Collapse
|
31
|
Langenbacher AD, Nguyen CT, Cavanaugh AM, Huang J, Lu F, Chen JN. The PAF1 complex differentially regulates cardiomyocyte specification. Dev Biol 2011; 353:19-28. [PMID: 21338598 DOI: 10.1016/j.ydbio.2011.02.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 02/07/2011] [Accepted: 02/11/2011] [Indexed: 11/28/2022]
Abstract
The specification of an appropriate number of cardiomyocytes from the lateral plate mesoderm requires a careful balance of both positive and negative regulatory signals. To identify new regulators of cardiac specification, we performed a phenotype-driven ENU mutagenesis forward genetic screen in zebrafish. In our genetic screen we identified a zebrafish ctr9 mutant with a dramatic reduction in myocardial cell number as well as later defects in primitive heart tube elongation and atrioventricular boundary patterning. Ctr9, together with Paf1, Cdc73, Rtf1 and Leo1, constitute the RNA polymerase II associated protein complex, PAF1. We demonstrate that the PAF1 complex (PAF1C) is structurally conserved among zebrafish and other metazoans and that loss of any one of the components of the PAF1C results in abnormal development of the atrioventricular boundary of the heart. However, Ctr9, Cdc73, Paf1 and Rtf1, but not Leo1, are required for the specification of an appropriate number of cardiomyocytes and elongation of the heart tube. Interestingly, loss of Rtf1 function produced the most severe defects, resulting in a nearly complete absence of cardiac precursors. Based on gene expression analyses and transplantation studies, we found that the PAF1C regulates the developmental potential of the lateral plate mesoderm and is required cell autonomously for the specification of cardiac precursors. Our findings demonstrate critical but differential requirements for PAF1C components in zebrafish cardiac specification and heart morphogenesis.
Collapse
Affiliation(s)
- Adam D Langenbacher
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
The myocardium of the heart is composed of multiple highly specialized myocardial lineages, including those of the ventricular and atrial myocardium, and the specialized conduction system. Specification and maturation of each of these lineages during heart development is a highly ordered, ongoing process involving multiple signaling pathways and their intersection with transcriptional regulatory networks. Here, we attempt to summarize and compare much of what we know about specification and maturation of myocardial lineages from studies in several different vertebrate model systems. To date, most research has focused on early specification, and although there is still more to learn about early specification, less is known about factors that promote subsequent maturation of myocardial lineages required to build the functioning adult heart.
Collapse
Affiliation(s)
- Sylvia M Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Medicine, University of California San Diego, 9500 Gilman Dr, La Jolla CA 92093, USA.
| | | | | | | |
Collapse
|
33
|
Affiliation(s)
- Michela Noseda
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| | - Tessa Peterkin
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| | - Filipa C. Simões
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| | - Roger Patient
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| | - Michael D. Schneider
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| |
Collapse
|
34
|
Chopra SS, Stroud DM, Watanabe H, Bennett JS, Burns CG, Wells KS, Yang T, Zhong TP, Roden DM. Voltage-gated sodium channels are required for heart development in zebrafish. Circ Res 2010; 106:1342-50. [PMID: 20339120 DOI: 10.1161/circresaha.109.213132] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
RATIONALE Voltage-gated sodium channels initiate action potentials in excitable tissues. Mice in which Scn5A (the predominant sodium channel gene in heart) has been knocked out die early in development with cardiac malformations by mechanisms which have yet to be determined. OBJECTIVE Here we addressed this question by investigating the role of cardiac sodium channels in zebrafish heart development. METHODS AND RESULTS Transcripts of the functionally-conserved Scn5a homologs scn5Laa and scn5Lab were detected in the gastrulating zebrafish embryo and subsequently in the embryonic myocardium. Antisense knockdown of either channel resulted in marked cardiac chamber dysmorphogenesis and perturbed looping. These abnormalities were associated with decreased expression of the myocardial precursor genes nkx2.5, gata4, and hand2 in anterior lateral mesoderm and significant deficits in the production of cardiomyocyte progenitors. These early defects did not appear to result from altered membrane electrophysiology, as prolonged pharmacological blockade of sodium current failed to phenocopy channel knockdown. Moreover, embryos grown in calcium channel blocker-containing medium had hearts that did not beat but developed normally. CONCLUSIONS These findings identify a novel and possibly nonelectrogenic role for cardiac sodium channels in heart development.
Collapse
Affiliation(s)
- Sameer S Chopra
- Department of Pharmacology, Vanderbilt University School of Medicine, 2215B Garland Ave, 1275 MRBIV Light Hall, Nashville, TN 37232-0575, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Samuel LJ, Latinkić BV. Early activation of FGF and nodal pathways mediates cardiac specification independently of Wnt/beta-catenin signaling. PLoS One 2009; 4:e7650. [PMID: 19862329 PMCID: PMC2763344 DOI: 10.1371/journal.pone.0007650] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Accepted: 10/07/2009] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Cardiac induction, the first step in heart development in vertebrate embryos, is thought to be initiated by anterior endoderm during gastrulation, but what the signals are and how they act is unknown. Several signaling pathways, including FGF, Nodal, BMP and Wnt have been implicated in cardiac specification, in both gain- and loss-of-function experiments. However, as these pathways regulate germ layer formation and patterning, their specific roles in cardiac induction have been difficult to define. METHODOLOGY/PRINCIPAL FINDINGS To investigate the mechanisms of cardiac induction directly we devised an assay based on conjugates of anterior endoderm from early gastrula stage Xenopus embryos as the inducing tissue and pluripotent ectodermal explants as the responding tissue. We show that the anterior endoderm produces a specific signal, as skeletal muscle is not induced. Cardiac inducing signal needs up to two hours of interaction with the responding tissue to produce an effect. While we found that the BMP pathway was not necessary, our results demonstrate that the FGF and Nodal pathways are essential for cardiogenesis. They were required only during the first hour of cardiogenesis, while sustained activation of ERK was required for at least four hours. Our results also show that transient early activation of the Wnt/beta-catenin pathway has no effect on cardiogenesis, while later activation of the pathway antagonizes cardiac differentiation. CONCLUSIONS/SIGNIFICANCE We have described an assay for investigating the mechanisms of cardiac induction by anterior endoderm. The assay was used to provide evidence for a direct, early and transient requirement of FGF and Nodal pathways. In addition, we demonstrate that Wnt/beta-catenin pathway plays no direct role in vertebrate cardiac specification, but needs to be suppressed just prior to differentiation.
Collapse
Affiliation(s)
- Lee J. Samuel
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Branko V. Latinkić
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
- * E-mail:
| |
Collapse
|
36
|
|
37
|
Differential requirement for BMP signaling in atrial and ventricular lineages establishes cardiac chamber proportionality. Dev Biol 2009; 328:472-82. [PMID: 19232521 DOI: 10.1016/j.ydbio.2009.02.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Revised: 02/04/2009] [Accepted: 02/06/2009] [Indexed: 11/23/2022]
Abstract
The function of an organ relies upon the proper relative proportions of its individual operational components. For example, effective embryonic circulation requires the appropriate relative sizes of each of the distinct pumps created by the atrial and ventricular cardiac chambers. Although the differences between atrial and ventricular cardiomyocytes are well established, little is known about the mechanisms regulating production of proportional numbers of each cell type. We find that mutation of the zebrafish type I BMP receptor gene alk8 causes reduction of atrial size without affecting the ventricle. Loss of atrial tissue is evident in the lateral mesoderm prior to heart tube formation and results from the inhibition of BMP signaling during cardiac progenitor specification stages. Comparison of the effects of decreased and increased BMP signaling further demonstrates that atrial cardiomyocyte production correlates with levels of BMP signaling while ventricular cardiomyocyte production is less susceptible to manipulation of BMP signaling. Additionally, mosaic analysis provides evidence for a cell-autonomous requirement for BMP signaling during cardiomyocyte formation and chamber fate assignment. Together, our studies uncover a new role for BMP signaling in the regulation of chamber size, supporting a model in which differential reception of cardiac inductive signals establishes chamber proportion.
Collapse
|
38
|
Kawahara A. Genetic dissection of cardiac progenitor migration. Inflamm Regen 2009. [DOI: 10.2492/inflammregen.29.324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
39
|
Taha MF, Valojerdi MR. Effect of bone morphogenetic protein-4 on cardiac differentiation from mouse embryonic stem cells in serum-free and low-serum media. Int J Cardiol 2007; 127:78-87. [PMID: 17714812 DOI: 10.1016/j.ijcard.2007.04.173] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2006] [Revised: 04/10/2007] [Accepted: 04/12/2007] [Indexed: 10/22/2022]
Abstract
In spite of previous reports, the precise role of bone morphogenetic proteins (BMPs) on cardiomyocyte differentiation, especially in the absence or presence of minimum amount of serum in culture medium is still unclear. So, the aim of the present study was to investigate the effect of BMP-4 on mouse embryonic stem cells (ESCs)-derived cardiomyocyte differentiation in serum-free and low-serum media. The mouse ESCs differentiation to cardiomyocytes was induced by embryoid bodies' (EBs') development through hanging drop, suspension and plating stages. Different models of differentiation were designed according to addition of fetal bovine serum (FBS) or knockout serum replacement (KoSR) to the medium of three stages. 10 ng/ml BMP-4 was added throughout the suspension period. Up to 30 days after plating, contraction and beating frequency were monitored and evaluated daily. The growth characteristics of cardiomyocytes were assessed by cardioactive drugs, immunocytochemistry, transmission electron microscopy (TEM) and reverse transcription-polymerase chain reaction (RT-PCR). In the complete absence of serum, neither control nor BMP-4 treated groups resulted in cardiac differentiation. Addition of FBS to hanging drop stage resulted in the appearance of beating cardiac clusters in some BMP-4 treated EBs. In the best designed differentiation model in which only hanging drop and the first 24 h of plating stage was carried out at the presence of FBS, the BMP-4 treatment resulted in cardiac differentiation in EBs characterized by positive immunostaining for the applied antibodies, chronotropic response to the cardioactive drugs and cardiac-specific genes expression at different developmental stages. These cardiomyocytes showed immature myofibrils and numerous intercellular junctions. In conclusion, BMP-4 is unable to induce cardiomyocyte differentiation from mouse ESCs in serum-free models, and at least small amount of FBS in hanging drop stage is necessary. Furthermore, serum factors are not strictly necessary after the initial activation, but they do favor a better differentiation of cardiomyocytes.
Collapse
Affiliation(s)
- Masoumeh Fakhr Taha
- Department of Anatomy, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | |
Collapse
|
40
|
Peterkin T, Gibson A, Patient R. Redundancy and evolution of GATA factor requirements in development of the myocardium. Dev Biol 2007; 311:623-35. [PMID: 17869240 PMCID: PMC2279743 DOI: 10.1016/j.ydbio.2007.08.018] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2007] [Revised: 08/01/2007] [Accepted: 08/07/2007] [Indexed: 11/26/2022]
Abstract
The transcription factors, GATA4, 5 and 6, recognize the same DNA sequence and are all expressed in the developing myocardium. However, knockout studies in the mouse have indicated that none of them are absolutely required for the specification of the myocardium. Here we present evidence for redundancy in this family for the first time. Using morpholinos in both Xenopus and zebrafish embryos, we show that GATA4 knockdown, for example, only affects cardiac marker expression in the absence of either GATA5 or GATA6. A similar situation pertains for GATA5 in Xenopus whereas, in zebrafish, GATA5 (faust) plays a major role in driving the myocardial programme. This requirement for GATA5 in zebrafish is for induction of the myocardium, in contrast to the GATA6 requirement in both species, which is for differentiation. This early role for GATA5 in zebrafish correlates with its earlier expression and with an earlier requirement for BMP signalling, suggesting that a mutual maintenance loop for GATA, BMP and Nkx expression is the evolutionarily conserved entity.
Collapse
Affiliation(s)
- Tessa Peterkin
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Abigail Gibson
- The Victor Chang Cardiac Research Institute, Level 6, 384 Victoria Street, Darlinghurst, NSW 2010, Sydney, Australia
| | - Roger Patient
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
- Corresponding author. Fax: +441865222501.
| |
Collapse
|
41
|
Miyasaka KY, Kida YS, Sato T, Minami M, Ogura T. Csrp1 regulates dynamic cell movements of the mesendoderm and cardiac mesoderm through interactions with Dishevelled and Diversin. Proc Natl Acad Sci U S A 2007; 104:11274-9. [PMID: 17592114 PMCID: PMC2040889 DOI: 10.1073/pnas.0702000104] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Zebrafish Csrp1 is a member of the cysteine- and glycine-rich protein (CSRP) family and is expressed in the mesendoderm and its derivatives. Csrp1 interacts with Dishevelled 2 (Dvl2) and Diversin (Div), which control cell morphology and other dynamic cell behaviors via the noncanonical Wnt and JNK pathways. When csrp1 message is knocked down, abnormal convergent extension cell movement is induced, resulting in severe deformities in midline structures. In addition, cardiac bifida is induced as a consequence of defects in cardiac mesoderm cell migration. Our data highlight Csrp1 as a key molecule of the noncanonical Wnt pathway, which orchestrates cell behaviors during dynamic morphogenetic movements of tissues and organs.
Collapse
Affiliation(s)
- Kota Y. Miyasaka
- Department of Developmental Neurobiology, Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo, Aoba, Sendai 980-8575, Japan; and Graduate School of Biological Sciences, Tohoku University, 2-1-1 Katahira, Aoba, Sendai 980-8577, Japan
| | - Yasuyuki S. Kida
- Department of Developmental Neurobiology, Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo, Aoba, Sendai 980-8575, Japan; and Graduate School of Biological Sciences, Tohoku University, 2-1-1 Katahira, Aoba, Sendai 980-8577, Japan
| | - Takayuki Sato
- Department of Developmental Neurobiology, Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo, Aoba, Sendai 980-8575, Japan; and Graduate School of Biological Sciences, Tohoku University, 2-1-1 Katahira, Aoba, Sendai 980-8577, Japan
| | - Mari Minami
- Department of Developmental Neurobiology, Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo, Aoba, Sendai 980-8575, Japan; and Graduate School of Biological Sciences, Tohoku University, 2-1-1 Katahira, Aoba, Sendai 980-8577, Japan
| | - Toshihiko Ogura
- Department of Developmental Neurobiology, Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo, Aoba, Sendai 980-8575, Japan; and Graduate School of Biological Sciences, Tohoku University, 2-1-1 Katahira, Aoba, Sendai 980-8577, Japan
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|
42
|
Tomescot A, Leschik J, Bellamy V, Dubois G, Messas E, Bruneval P, Desnos M, Hagège AA, Amit M, Itskovitz J, Menasché P, Pucéat M. Differentiation in vivo of cardiac committed human embryonic stem cells in postmyocardial infarcted rats. Stem Cells 2007; 25:2200-5. [PMID: 17540853 DOI: 10.1634/stemcells.2007-0133] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Human embryonic stem (HES) cells can give rise to cardiomyocytes in vitro. However, whether undifferentiated HES cells also feature a myocardial regenerative capacity after in vivo engraftment has not been established yet. We compared two HES cell lines (HUES-1 and I6) that were specified toward a cardiac lineage by exposure to bone morphogenetic protein-2 (BMP2) and SU5402, a fibroblast growth factor receptor inhibitor. Real-time polymerase chain reaction (PCR) revealed that the cardiogenic inductive factor turned on expression of mesodermal and cardiac genes (Tbx6, Isl1, FoxH1, Nkx2.5, Mef2c, and alpha-actin). Thirty immunosuppressed rats underwent coronary artery ligation and, 2 weeks later, were randomized and received in-scar injections of either culture medium (controls) or BMP2 (+/-SU5402)-treated HES cells. After 2 months, human cells were detected by anti-human lamin immunostaining, and their cardiomyocytic differentiation was evidenced by their expression of cardiac markers by reverse transcription-PCR and immunofluorescence using an anti-beta myosin antibody. No teratoma was observed in hearts or any other organ of the body. The ability of cardiac-specified HES cells to differentiate along the cardiomyogenic pathway following transplantation into infarcted myocardium raises the hope that these cells might become effective candidates for myocardial regeneration.
Collapse
Affiliation(s)
- André Tomescot
- Institut National de la Santé et de la Recherche Médicale/Evry University Unité Mixte de Recherche 861, I-Stem, Association Française contre les Myopathies, 5, rue Desbrières, Evry 91030, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Guzzo RM, Foley AC, Ibarra YM, Mercola M. Signaling Pathways in Embryonic Heart Induction. CARDIOVASCULAR DEVELOPMENT 2007. [DOI: 10.1016/s1574-3349(07)18005-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
44
|
Schoenebeck JJ, Yelon D. Illuminating cardiac development: Advances in imaging add new dimensions to the utility of zebrafish genetics. Semin Cell Dev Biol 2006; 18:27-35. [PMID: 17241801 PMCID: PMC1876688 DOI: 10.1016/j.semcdb.2006.12.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The use of the zebrafish as a model organism for the analysis of cardiac development is no longer proof-of-principle science. Over the last decade, the identification of a variety of zebrafish mutations and the subsequent cloning of mutated genes have revealed many critical regulators of cardiogenesis. More recently, increasingly sophisticated techniques for phenotypic characterization have facilitated analysis of the specific mechanisms by which key genes drive cardiac specification, morphogenesis, and function. Future enrichment of the arsenal of experimental strategies available for zebrafish should continue the yield of high returns from such a small source.
Collapse
Affiliation(s)
- Jeffrey J Schoenebeck
- Developmental Genetics Program and Department of Cell Biology, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | | |
Collapse
|
45
|
Taha MF, Valojerdi MR, Mowla SJ. Effect of bone morphogenetic protein-4 (BMP-4) on cardiomyocyte differentiation from mouse embryonic stem cell. Int J Cardiol 2006; 120:92-101. [PMID: 17156864 DOI: 10.1016/j.ijcard.2006.08.118] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2006] [Revised: 08/07/2006] [Accepted: 08/12/2006] [Indexed: 11/25/2022]
Abstract
The present study was designed to evaluate the effect of BMP-4 on mouse embryonic stem cells (ESCs)-derived cardiomyocyte. Cardiac differentiation of the mouse ESCs was initiated by embryoid bodies (EBs) formation in hanging drops, transfer of EBs to the suspension culture and then plating onto gelatin-coated tissue culture plates. BMP-4 was added to culture medium throughout the suspension period. Cultures were observed daily with an inverted microscope for the appearance of contracting clusters. At the early, intermediate and terminal stages of differentiation, the choronotropic responses of cardiomyocytes to cardioactive drugs were assessed, and the cardiomyocytes immunostained for cardiac troponin I, desmin, alpha-actinin and nebulin. The contracting clusters were isolated for ultrastructural evaluation, at day 14 after plating. Moreover, total RNA extracted from contracting EBs of early and terminal stages of differentiation were examined for oct-4, alpha- and beta-myosin heavy chain, myosin light chain-2V and atrial natriuretic factor expression. The BMP-4 treatment resulted in a decrease in the percent of beating EBs and the percent of developing cardiomyocytes per EBs. As a whole, the chronotropic responses of beating cardiac clusters to cardioactive drugs in control group were better than BMP-4 treated group. The cardiomyocytes of both groups were positive immunostained for applied antibodies except for nebulin. Moreover, in the BMP-4 treated group, the ultrastructural characteristics and cardiac-specific genes expression were all retarded in the terminal stage of cardiomyocytes development. In conclusion, BMP-4 had an inhibitory effect on cardiomyocyte differentiation from the mouse ESCs in terms of ultrastructural characteristics, genes expression and functional properties.
Collapse
Affiliation(s)
- Masoumeh Fakhr Taha
- Department of Anatomy, School of Medical Sciences, Tarbiat Modarres University, Tehran, Iran
| | | | | |
Collapse
|
46
|
Foley AC, Korol O, Timmer AM, Mercola M. Multiple functions of Cerberus cooperate to induce heart downstream of Nodal. Dev Biol 2006; 303:57-65. [PMID: 17123501 PMCID: PMC1855199 DOI: 10.1016/j.ydbio.2006.10.033] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Revised: 10/05/2006] [Accepted: 10/22/2006] [Indexed: 11/25/2022]
Abstract
The TGFbeta family member Nodal has been implicated in heart induction through misexpression of a dominant negative version of the type I Nodal receptor (Alk4) and targeted deletion of the co-receptor Cripto in murine ESCs and mouse embryos; however, whether Nodal acts directly or indirectly to induce heart tissue or interacts with other signaling molecules or pathways remained unclear. Here we present Xenopus embryological studies demonstrating an unforeseen role for the DAN family protein Cerberus within presumptive foregut endoderm as essential for differentiation of cardiac mesoderm in response to Nodal. Ectopic activation of Nodal signaling in non-cardiogenic ventroposterior mesendoderm, either by misexpression of the Nodal homologue XNr1 together with Cripto or by a constitutively active Alk4 (caAlk4), induced both cardiac markers and Cerberus. Mosaic lineage tracing studies revealed that Nodal/Cripto and caAlk4 induced cardiac markers cell non-autonomously, thus supporting the idea that Cerberus or another diffusible factor is an essential mediator of Nodal-induced cardiogenesis. Cerberus alone was found sufficient to initiate cardiogenesis at a distance from its site of synthesis. Conversely, morpholino-mediated specific knockdown of Cerberus reduced both endogenous cardiomyogenesis and ectopic heart induction resulting from misactivation of Nodal/Cripto signaling. Since the specific knockdown of Cerberus did not abrogate heart induction by the Wnt antagonist Dkk1, Nodal/Cripto and Wnt antagonists appear to initiate cardiogenesis through distinct pathways. This idea was further supported by the combinatorial effect of morpholino-medicated knockdown of Cerberus and Hex, which is required for Dkk1-induced cardiogenesis, and the differential roles of essential downstream effectors: Nodal pathway activation did not induce the transcriptional repressor Hex while Dkk-1 did not induce Cerberus. These studies demonstrated that cardiogenesis in mesoderm depends on Nodal-mediated induction of Cerberus in underlying endoderm, and that this pathway functions in a pathway parallel to cardiogenesis initiated through the induction of Hex by Wnt antagonists. Both pathways operate in endoderm to initiate cardiogenesis in overlying mesoderm.
Collapse
Affiliation(s)
| | | | | | - Mark Mercola
- *author for correspondence, E-mail: , Telephone: (858) 795-5242, Fax: (858) 713 6274
| |
Collapse
|
47
|
Inui M, Fukui A, Ito Y, Asashima M. Xapelin and Xmsr are required for cardiovascular development in Xenopus laevis. Dev Biol 2006; 298:188-200. [PMID: 16876154 DOI: 10.1016/j.ydbio.2006.06.028] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2006] [Revised: 06/11/2006] [Accepted: 06/15/2006] [Indexed: 11/25/2022]
Abstract
The cardiovascular development is the elaborate process, and despite the extensive studies, the mechanisms underlying endothelial, hematopoietic, and cardiac developments, as well as the interrelation between these processes, are not fully understood. In this study, we demonstrated that Xenopus apelin and Xmsr play pivotal roles in cardiovascular development. Apelin is a recently identified ligand for an orphan G-protein-coupled receptor APJ and is involved in fluid homeostasis in mammals. Xenopus preproapelin (Xpreproapelin) was isolated and its mRNA localized to the region around the presumptive blood vessels, which are overlapping or adjacent to those expressing Xmsr, the Xenopus homologue of APJ. Overexpression of Xpreproapelin disorganized the expression of the endothelial precursor cell marker XlFli and the hematopoietic precursor cell marker SCL at the neurula, whereas embryos injected with morpholino antisense oligonucleotides for Xapelin and Xmsr displayed attenuated expression of Tie2, alpha-globin, XPOX2, and cTnI, markers of endothelium, erythrocytes, myeloid cells, and cardiomyocytes, respectively. XlFli morpholino had similar effects to Xapelin and Xmsr morpholinos on cardiac differentiation, suggesting an unexpected potential relationship between the endothelium and cardiac differentiation. Forced expression of constitutive active G alpha i rescued the phenotypes of Xmsr morpholino-injected embryos, indicating that the i/o type of G protein alpha subunit acts downstream of Xmsr.
Collapse
Affiliation(s)
- Masafumi Inui
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | | | | | | |
Collapse
|
48
|
Moreno-Rodriguez RA, Krug EL, Reyes L, Villavicencio L, Mjaatvedt CH, Markwald RR. Bidirectional fusion of the heart-forming fields in the developing chick embryo. Dev Dyn 2006; 235:191-202. [PMID: 16252277 PMCID: PMC1855217 DOI: 10.1002/dvdy.20601] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
It is generally thought that the early pre-tubular chick heart is formed by fusion of the anterior or cephalic limits of the paired cardiogenic fields. However, this study shows that the heart fields initially fuse at their midpoint to form a transitory "butterfly"-shaped, cardiogenic structure. Fusion then progresses bi-directionally along the longitudinal axis in both cranial and caudal directions. Using in vivo labeling, we demonstrate that cells along the ventral fusion line are highly motile, crossing future primitive segments. We found that mesoderm cells migrated cephalically from the unfused tips of the anterior/cephalic wings into the head mesenchyme in the region that has been called the secondary heart field. Perturbing the anterior/cranial fusion results in formation of a bi-conal heart. A theoretical role of the ventral fusion line acting as a "heart organizer" and its role in cardia bifida is discussed.
Collapse
Affiliation(s)
- R A Moreno-Rodriguez
- Department of Cell Biology and Anatomy, Medical University of South Carolina, Charleston, South Carolina 29425, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Pucéat M. Les cellules souches embryonnaires : Du développement myocardique à la médecine régénératrice. Med Sci (Paris) 2005; 21:1076-82. [PMID: 16324649 DOI: 10.1051/medsci/200521121076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Embryonic stem cells are capable to recapitulate the first stages of myocardial development. Using mouse embryonic stem cells, transcriptional networks specifying the cardiac fate can be delineated. Furthermore, using members of the TGFbeta superfamily to commit mouse ES cells toward a cardiac lineage, recent studies showed that ESC-derived cardiomyocytes were capable to repair post-infarcted myocardium of small and large animals. The next challenges are to validate such results using human ESCs in order to better comprehend cardiac congenital diseases and to foresee a cell therapy of heart failure. double dagger.
Collapse
Affiliation(s)
- Michel Pucéat
- CNRS FRE2593, Centre de Recherches de Biochimie macromoléculaire, 1919, route de Mende, 34293 Montpellier Cedex, France.
| |
Collapse
|
50
|
Brewer AC, Alexandrovich A, Mjaatvedt CH, Shah AM, Patient RK, Pizzey JA. GATA factors lie upstream of Nkx 2.5 in the transcriptional regulatory cascade that effects cardiogenesis. Stem Cells Dev 2005; 14:425-39. [PMID: 16137232 DOI: 10.1089/scd.2005.14.425] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Members of the GATA-4, -5, and -6 subfamily of transcription factors are co-expressed with the homeoprotein Nkx 2.5 in the precardiac mesoderm during the earliest stages of its specification and are known to be important determinants of cardiac gene expression. Ample evidence suggests that GATA factors and Nkx 2.5 cross-regulate each other's expression; however, the temporal order of the expression of these transcription factors in vivo remains unresolved, and thus precise definition of the role of the products of the genes they transcribe in early development has been difficult to assess. We employed P19 CL6 mouse embryonic carcinoma cells as a model to investigate this problem, because these cells, like embryonic stem cells, can be induced to differentiate along multiple lineages. Here we demonstrate that when P19 CL6 cells are induced to differentiate to a cardiogenic lineage, the expression of GATA-4 and GATA-6 is up-regulated prior to the transcriptional activation of Nkx 2.5. Moreover, over-expression of GATA-4 or -6 at the time of Nkx 2.5 induction results in a significant up-regulation of endogenous Nkx 2.5 transcription. Finally, it is known that a Nkx-dependent enhancer is necessary for GATA-6 expression within cardiomyocytes of the developing mouse embryo. We demonstrate that within undifferentiated P19 CL6 cells, GATA-6 expression is subject to active repression by a novel upstream element that possesses binding sites for factors involved in transcriptional repression that are conserved between mammalian species.
Collapse
Affiliation(s)
- Alison C Brewer
- Department of Cardiology, King's College Hospital, London SE5 9RS, UK
| | | | | | | | | | | |
Collapse
|